1
|
D'Orsi G, Palladini M, Mazza MG, Rovere-Querini P, Scalabrini A, Benedetti F. A novel analysis of interoceptive underpinnings of anxious psychopathology in COVID-19 survivors. Behav Brain Res 2025; 476:115275. [PMID: 39332641 DOI: 10.1016/j.bbr.2024.115275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/16/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
INTRODUCTION SARS-CoV-2 affects brain, body, and their interchange. We investigated interoceptive mechanisms in COVID-19 survivors focusing on their potential link with psychopathology and inflammatory biomarkers. METHODS We assessed interoceptive accuracy (IAc) and time-perceiving (TA) skills of 57 COVID-19 survivors one month after hospital discharge through, respectively, a heartbeats perception task and a time duration task. Each participant was assessed about his interoceptive awareness (IAw) through Multidimensional Assessment of Interoceptive Awareness questionnaire (MAIA) and then, screened for post-traumatic (Impact of Events Scale - IES-R), anxious (State-Trait Anxiety Inventory - STAI-Y1) and depressive (Zung Self-Rating Depression Scale - ZSDS; Beck Depression Inventory - BDI-13) symptoms. Biomarkers of inflammation (platelet count, PC; mean platelet volume, MPV and systemic immune-inflammation index, SII) were obtained in a subsample of 40 survivors by a blood sampling conducted at admission and discharge time from the hospital. Correlational, GLM, GLMZ, and mediation analyses were performed. RESULTS IAc did not correlate with TA confirming the reliability of interoceptive measure. IAc positively predicts MAIA's Trusting subscale and negatively predicts anxious psychopathology which fully mediates the effect of IAc on Trusting.PC at hospital admission predicts anxiety at one month after recovery. Again, a higher decrease of SII during hospitalization predicts higher IAc skill and lower anxiety state at one month. The link between SII change and anxiety is fully mediated by IAc. CONCLUSIONS Our results unveil a potential key role of interoception and brain-body interchange in the exacerbation and maintenance of anxiety psychopathology in COVID-19 survivors.
Collapse
Affiliation(s)
- Greta D'Orsi
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy.
| | - Mariagrazia Palladini
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy.
| | - Mario Gennaro Mazza
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy.
| | - Patrizia Rovere-Querini
- University Vita-Salute San Raffaele, Milano, Italy; Unit of Innate Immunity and Tissue Remodeling, Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Hospital, Milano, Italy.
| | - Andrea Scalabrini
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy; Department of Human and Social Sciences University of Bergamo, Bergamo, Italy.
| | - Francesco Benedetti
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy.
| |
Collapse
|
2
|
Bianconi V, Mannarino MR, Figorilli F, Ricciutelli F, De Carlo S, Zullo V, Corba M, Sahebkar A, Greco A, Lombardini R, Paltriccia R, Pirro M. Treatment with proton pump inhibitors is associated with secondary bacterial infections and sepsis in patients with COVID-19: a retrospective analysis of their joint impact on in-hospital prognosis. Ann Med 2024; 56:2399761. [PMID: 39475004 DOI: 10.1080/07853890.2024.2399761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/29/2024] [Accepted: 04/27/2024] [Indexed: 11/10/2024] Open
Abstract
Introduction and objectives. Secondary bacterial infections (SBIs) contribute to worse in-hospital outcomes in patients with Coronavirus disease 2019 (COVID-19). Treatment with proton pump inhibitors (PPIs) is associated with an increased risk of bacterial infections in different clinical settings. However, the association between PPI treatment prior to hospital admission and the occurrence of either SBIs or secondary bacterial sepsis (SBS) as well as their joint impact on clinical outcomes of patients hospitalized for COVID-19 are not clarified. Patients and methods. We retrospectively analyzed preadmission PPI use, in-hospital occurrence of SBIs and SBS, and in-hospital outcomes of a cohort of patients hospitalized for COVID-19. Results. Among 1087 patients, 447 (41%) were on PPI treatment prior to hospital admission. During the hospital stay, 197 (18%) and 223 (20%) patients were diagnosed with SBIs and SBS, respectively. The composite endpoint of intensive care unit (ICU) admission/in-hospital death was met by 214 (20%) patients. Preadmission PPI treatment was independently associated with up to a 2.1-fold and 1.7-fold increased risk of SBIs and SBS, respectively. The occurrence of SBS was independently associated with up to a 2.2-fold increased risk of ICU admission/in-hospital death. A significant preadmission PPI treatment x SBS interaction emerged in predicting ICU admission/in-hospital death (F = 5.221, pinteraction = 0.023). Conclusions. PPI treatment prior to hospital admission for COVID-19 is associated with an increased risk of SBIs and SBS. In addition, it interacts with SBS in predicting in-hospital prognosis. An appropriate use of PPIs may attenuate the risk of adverse clinical outcomes during hospitalization for COVID-19.
Collapse
Affiliation(s)
- Vanessa Bianconi
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Massimo R Mannarino
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Filippo Figorilli
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Federica Ricciutelli
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Stefania De Carlo
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Valentina Zullo
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Martina Corba
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alessia Greco
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Rita Lombardini
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Rita Paltriccia
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Matteo Pirro
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
3
|
Teng L, Chang G, Song X, Zhang M, Han Y, Chang W, Shen Z. Construction and validation of a risk model of proteinuria in patients with omicron COVID-19: retrospective cohort study. Ren Fail 2024; 46:2365979. [PMID: 39108141 PMCID: PMC11308959 DOI: 10.1080/0886022x.2024.2365979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND To explore the risk factors of proteinuria in Omicron variant patients and to construct and verify the risk predictive model. METHODS 1091 Omicron patients who were hospitalized from August 2022 to November 2022 at Tianjin First Central Hospital were defined as the derivation cohort. 306 Omicron patients who were hospitalized from January 2022 to March 2022 at the same hospital were defined as the validation cohort. The risk factors of proteinuria in derivation cohort were screened by univariate and multivariate logistic regression analysis, and proteinuria predicting scoring system was constructed and the receiver operating characteristic(ROC)curve was drawn to test the prediction ability. The proteinuria risk model was externally validated in validation cohort. RESULTS 7 factors including comorbidities, blood urea nitrogen (BUN), serum sodium (Na), uric acid (UA), C reactive protein (CRP) and vaccine dosages were included to construct a risk predictive model. The score ranged from -5 to 16. The area under the ROC curve(AUC) of the model was 0.8326(95% CI 0.7816 to 0.8835, p < 0.0001). Similarly to that observed in derivation cohort, the AUC is 0.833(95% CI 0.7808 to 0.9002, p < 0.0001), which verified good prediction ability and diagnostic accuracy in validation cohort. CONCLUSIONS The risk model of proteinuria after Omicron infection had better assessing efficiency which could provide reference for clinical prediction of the risk of proteinuria in Omicron patients.
Collapse
Affiliation(s)
- Lanbo Teng
- Department of Nephrology, Tianjin First Central Hospital, Nankai University, Tianjin, China
- National Health Commission (NHC) Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Ge Chang
- Department of Clinical Medicine, Tianjin Medical University, Tianjin, China
| | - Xinyuan Song
- Department of Nephrology, Tianjin First Central Hospital, Nankai University, Tianjin, China
- National Health Commission (NHC) Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Miaomiao Zhang
- Department of Nephrology, Tianjin First Central Hospital, Nankai University, Tianjin, China
- National Health Commission (NHC) Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Yingying Han
- Department of Nephrology, Tianjin First Central Hospital, Nankai University, Tianjin, China
- National Health Commission (NHC) Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Wenxiu Chang
- Department of Nephrology, Tianjin First Central Hospital, Nankai University, Tianjin, China
- National Health Commission (NHC) Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Zhongyang Shen
- National Health Commission (NHC) Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin, China
| |
Collapse
|
4
|
Mohamad NA, Yong MH, Mohd Khialdin S, Bastion MLC. Case report: Acute macular neuroretinopathy post-COVID-19 infection. Optom Vis Sci 2024:00006324-990000000-00239. [PMID: 39514396 DOI: 10.1097/opx.0000000000002194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
SIGNIFICANCE Acute macular neuroretinopathy (AMN) is a rare disease that causes transient or permanent visual disturbance. The exact etiology remains unknown, but vascular compromise of the deep retinal capillary plexus was postulated as the main mechanism. Retinal vascular event post-coronavirus disease 2019 (COVID-19) infection is recently highlighted during the pandemic, which includes AMN. PURPOSE To report a case of AMN post-COVID-19 infection. CASE REPORT A 24-year-old Indian woman presented with acute-onset painless bilateral central scotoma for a day. The symptom was preceded by a history of COVID-19 infection 3 weeks prior. Ocular examination revealed a near-normal visual acuity for both eyes. Fundus examination showed bilateral dull foveal reflex with mild scattered cotton wool spot and vascular tortuosity. Optical coherence tomography macula revealed a distinct short hyperreflective band involving the outer plexiform and outer nuclear layers nasal to the fovea. The Bjerrum perimetry test revealed central scotoma temporal to the fixation. Optical coherence tomography lesions and scotomas are identical and congruous in both eyes. Serial fundus photographs are captured showing the evolving changes of near-normal macula to pigmented wedge-shaped petaloid lesions around the fovea. The patient was diagnosed as bilateral AMN and treated with oral prednisolone. On subsequent follow-up, the central scotoma improved. CONCLUSIONS This case illustrates a clear temporal and possible causal relationship of COVID-19 infection with AMN. Further studies and data are required to justify its association, but the rising cases of AMN shall be anticipated as COVID-19 infections have become endemic worldwide.
Collapse
Affiliation(s)
- Nur Ain Mohamad
- Department of Ophthalmology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Malaysia
| | - Meng Hsien Yong
- Department of Ophthalmology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Malaysia
| | - Safinaz Mohd Khialdin
- Department of Ophthalmology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Malaysia
| | | |
Collapse
|
5
|
Chen X, Jiang B, Gu Y, Yue Z, Liu Y, Lei Z, Yang G, Deng M, Zhang X, Luo Z, Li Y, Zhang Q, Zhang X, Wu J, Huang C, Pan P, Zhou F, Wang N. SARS-CoV-2 nucleocapsid protein interaction with YBX1 displays oncolytic properties through PKM mRNA destabilization. Mol Cancer 2024; 23:248. [PMID: 39506849 PMCID: PMC11539619 DOI: 10.1186/s12943-024-02153-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 10/10/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND SARS-CoV-2, a highly contagious coronavirus, is responsible for the global pandemic of COVID-19 in 2019. Currently, it remains uncertain whether SARS-CoV-2 possesses oncogenic or oncolytic potential in influencing tumor progression. Therefore, it is important to evaluate the clinical and functional role of SARS-CoV-2 on tumor progression. METHODS Here, we integrated bioinformatic analysis of COVID-19 RNA-seq data from the GEO database and performed functional studies to explore the regulatory role of SARS-CoV-2 in solid tumor progression, including lung, colon, kidney and liver cancer. RESULTS Our results demonstrate that infection with SARS-CoV-2 is associated with a decreased expression of genes associated with cancer proliferation and metastasis in lung tissues from patients diagnosed with COVID-19. Several cancer proliferation or metastasis related genes were frequently downregulated in SARS-CoV-2 infected intestinal organoids and human colon carcinoma cells. In vivo and in vitro studies revealed that SARS-CoV-2 nucleocapsid (N) protein inhibits colon and kidney tumor growth and metastasis through the N-terminal (NTD) and the C-terminal domain (CTD). The molecular mechanism indicates that the N protein of SARS-CoV-2 interacts with YBX1, resulting in the recruitment of PKM mRNA into stress granules mediated by G3BP1. This process ultimately destabilizes PKM expression and suppresses glycolysis. CONCLUSION Our study reveals a new function of SARS-CoV-2 nucleocapsid protein on tumor progression.
Collapse
Affiliation(s)
- Xin Chen
- Institute of Medical Microbiology, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Ministry of Education, Guangzhou, 510632, China
| | - Baohong Jiang
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yu Gu
- Institute of Medical Microbiology, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Ministry of Education, Guangzhou, 510632, China
| | - Zhaoyang Yue
- Institute of Medical Microbiology, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Ministry of Education, Guangzhou, 510632, China
| | - Ying Liu
- Institute of Medical Microbiology, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Ministry of Education, Guangzhou, 510632, China
| | - Zhiwei Lei
- Institute of Medical Microbiology, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Ministry of Education, Guangzhou, 510632, China
- Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511500, China
| | - Ge Yang
- Foshan Institute of Medical Microbiology, Foshan, 528315, China
- Section of Cellular and Molecular Biology, The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| | - Minhua Deng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Xuelong Zhang
- Institute of Medical Microbiology, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Ministry of Education, Guangzhou, 510632, China
| | - Zhen Luo
- Institute of Medical Microbiology, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Ministry of Education, Guangzhou, 510632, China
| | - Yongkui Li
- Institute of Medical Microbiology, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Ministry of Education, Guangzhou, 510632, China
| | - Qiwei Zhang
- Institute of Medical Microbiology, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Ministry of Education, Guangzhou, 510632, China
| | - Xuepei Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Jianguo Wu
- Institute of Medical Microbiology, Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Ministry of Education, Guangzhou, 510632, China
| | - Chunyu Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
- Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China.
| | - Pan Pan
- School of Basic Medical Science, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China.
- The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China.
| | - Fangjian Zhou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Ning Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450003, China.
| |
Collapse
|
6
|
Song HW, Jo HY, Kim SC, Choi SS. Immunopathological markers and cell types linked to COVID-19 symptom manifestation. BMC Infect Dis 2024; 24:1237. [PMID: 39497098 PMCID: PMC11533414 DOI: 10.1186/s12879-024-10139-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/28/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND Numerous studies have investigated the molecular properties that contribute to the symptoms of COVID-19, such as the virus's genetic makeup, its replication mechanisms, and how it interacts with host cells. However, identifying the immunopathological properties, such as the immune system's response, cytokine levels, and the presence of specific biomarkers, that are associated with the severity of the infection remains crucial for developing effective treatments and preventions. METHODS We analyzed blood protein factor profiles from 420 individuals to identify features differentiating between test-negative healthy, asymptomatic, and symptomatic individuals using statistical comparison and the least absolute shrinkage and selection operator (i.e., LASSO) algorithm. Additionally, we examined single-cell RNA sequencing data from 141 individuals to identify specific cell types associated with the COVID-19 symptoms. RESULTS Healthy individuals who tested negative had distinct blood protein factor levels compared to asymptomatic individuals. We identified two key protein factors, Serpin A10 and Complement C9, that differentiate between asymptomatic and symptomatic patients. Symptomatic patients showed lower levels of CD4+ T naïve, CD4+ T effector & memory, and CD8+ T naïve cells, along with higher levels of CD14+ classical monocytes compared to asymptomatic patients. Additionally, CD16+ non-classical monocytes, major producers of C1QA/B/C, appeared to contribute to the observed Complement C9 levels. CONCLUSIONS These findings advance our understanding of the immunopathological mechanisms underlying COVID-19 and may inform the development of targeted therapies and preventative measures. Future research should focus on further elucidating these mechanisms and exploring their potential clinical applications in managing COVID-19 severity.
Collapse
Affiliation(s)
- Ha Won Song
- Division of Biomedical Convergence, College of Biomedical Science, Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon, 24341, Korea
| | - Hye-Yeong Jo
- Division of Healthcare and Artificial Intelligence, Department of Precision Medicine, Korea National Institute of Health, Osong, 28159, Korea
| | - Sang Cheol Kim
- Division of Healthcare and Artificial Intelligence, Department of Precision Medicine, Korea National Institute of Health, Osong, 28159, Korea
| | - Sun Shim Choi
- Division of Biomedical Convergence, College of Biomedical Science, Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon, 24341, Korea.
| |
Collapse
|
7
|
Kim HJ, Kim MH, Choi MG, Chun EM. Psychiatric adverse events following COVID-19 vaccination: a population-based cohort study in Seoul, South Korea. Mol Psychiatry 2024; 29:3635-3643. [PMID: 38834668 PMCID: PMC11541197 DOI: 10.1038/s41380-024-02627-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/06/2024]
Abstract
Evidence has suggested an increased risk of psychiatric manifestations following viral infections including coronavirus disease-2019 (COVID-19). However, psychiatric adverse events (AEs) after COVID-19 vaccination, which were documented in case reports and case series, remain unclear. This study is aimed to investigate the psychiatric AEs after COVID-19 vaccination from a large population-based cohort in Seoul, South Korea. We recruited 50% of the Seoul-resident population randomly selected from the Korean National Health Insurance Service (KNHIS) claims database on 1, January, 2021. The included participants (n = 2,027,353) from the Korean National Health Insurance Service claims database were divided into two groups according to COVID-19 vaccination. The cumulative incidences per 10,000 of psychiatric AEs were assessed on one week, two weeks, one month, and three months after COVID-19 vaccination. Hazard ratios (HRs) and 95% Confidence interval (CIs) of psychiatric AEs were measured for the vaccinated population. The cumulative incidence of depression, anxiety, dissociative, stress-related, and somatoform disorders, sleep disorders, and sexual disorders at three months following COVID-19 vaccination were higher in the vaccination group than no vaccination group. However, schizophrenia and bipolar disorders showed lower cumulative incidence in the vaccination group than in the non-vaccinated group. Depression (HR [95% CI] = 1.683 [1.520-1.863]), anxiety, dissociative, stress-related, and somatoform disorders (HR [95% CI] = 1.439 [1.322-1.568]), and sleep disorders (HR [95% CI] = 1.934 [1.738-2.152]) showed increased risks after COVID-19 vaccination, whereas the risks of schizophrenia (HR [95% CI] = 0.231 [0.164-0.326]) and bipolar disorder (HR [95% CI] = 0.672 [0.470-0.962]). COVID-19 vaccination increased the risks of depression, anxiety, dissociative, stress-related, and somatoform disorders, and sleep disorders while reducing the risk of schizophrenia and bipolar disorder. Therefore, special cautions are necessary for administering additional COVID-19 vaccinations to populations vulnerable to psychiatric AEs.
Collapse
Affiliation(s)
- Hong Jin Kim
- Department of Orthopedic Surgery, Inje University Sanggye Paik Hospital, College of Medicine, Inje University, Seoul, Republic of Korea
| | - Min-Ho Kim
- Informatization Department, Ewha Womans University Seoul Hospital, Seoul, Republic of Korea
| | - Myeong Geun Choi
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, School of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Eun Mi Chun
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, School of Medicine, Ewha Womans University, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Khanal R, Heinen N, Bogomolova A, Meister TL, Herrmann ST, Westhoven S, Nocke MK, Todt D, Jockenhövel F, Klein IM, Hartmann L, Vondran FWR, Steinmann E, Zimmer G, Ott M, Brown RJP, Sharma AD, Pfaender S. MicroRNAs modulate SARS-CoV-2 infection of primary human hepatocytes by regulating the entry factors ACE2 and TMPRSS2. Liver Int 2024; 44:2983-2995. [PMID: 39175256 DOI: 10.1111/liv.16079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 08/10/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND AND AIMS Severe acute respiratory syndrome coronavirus (SARS-CoV-2) preferentially infects the respiratory tract; however, several studies have implicated a multi-organ involvement. Hepatic dysfunctions caused by SARS-CoV-2 infection have been increasingly recognized and described to correlate with disease severity. To elucidate molecular factors that could contribute towards hepatic infection, we concentrated on microRNAs (miRNAs), a class of small non-coding RNAs that modulate various cellular processes and which are reported to be differentially regulated during liver injury. We aimed to study the infection of primary human hepatocytes (PHH) with SARS-CoV-2 and to evaluate the potential of miRNAs for modulating viral infection. METHODS We analysed liver autopsies from a coronavirus disease 19 (COVID-19)-positive cohort for the presence of viral RNA using Nanopore sequencing. PHH were used for the infection with SARS-CoV-2. The candidate miRNAs targeting angiotensin converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2) were identified using in silico approaches. To discover the potential regulatory mechanism, transfection experiments, qRT-PCRs, western blots and luciferase reporter assays were performed. RESULTS We could detect SARS-CoV-2 RNA in COVID-19-positive liver autopsies. We show that PHH express ACE2 and TMPRSS2 and can be readily infected with SARS-CoV-2, resulting in robust replication. Transfection of selected miRNA mimics reduced SARS-CoV-2 receptor expression and SARS-CoV-2 burden in PHH. In silico and biochemical analyses supported a potential direct binding of miR-141-3p to the SARS-CoV-2 genome. CONCLUSION We confirm that PHH are susceptible to SARS-CoV-2 infection and demonstrate selected miRNAs targeting SARS-CoV-2 entry factors and/or the viral genome reduce viral loads. These data provide novel insights into hepatic susceptibility to SARS-CoV-2 and associated dysfunctions in COVID-19.
Collapse
Affiliation(s)
- Rajendra Khanal
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
- Research Group Liver Regeneration & RNA Therapeutics, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Natalie Heinen
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Alexandra Bogomolova
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
- Research Group Liver Regeneration & RNA Therapeutics, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Toni L Meister
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
- Institute for Infection Research and Vaccine Development (IIRVD), Centre for Internal Medicine, University Medical Centre Hamburg-Eppendorf (UKE), Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Centre for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Simon T Herrmann
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
- Research Unit Emerging Viruses, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Saskia Westhoven
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
- Research Unit Emerging Viruses, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Maximilian K Nocke
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Daniel Todt
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
- European Virus Bioinformatics Center (EVBC), Jena, Germany
| | - Freya Jockenhövel
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Isabel M Klein
- Tissue Bank of the German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Laura Hartmann
- Tissue Bank of the German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Florian W R Vondran
- Department of General, Visceral, Pediatric and Transplant Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Eike Steinmann
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Gert Zimmer
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Michael Ott
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Richard J P Brown
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Amar Deep Sharma
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
- Research Group Liver Regeneration & RNA Therapeutics, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Stephanie Pfaender
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
- Research Unit Emerging Viruses, Leibniz Institute of Virology (LIV), Hamburg, Germany
- University of Lübeck, Lübeck, Germany
| |
Collapse
|
9
|
Suleiman A, Munoz-Acuna R, Redaelli S, Ahrens E, Tartler TM, Ashrafian S, Hashish MM, Santarisi A, Chen G, Riedel S, Talmor D, Baedorf Kassis EN, Schaefer MS, Goodspeed V. Previous Coronavirus Disease-2019 Infection and Lung Mechanics in Surgical Patients: A Hospital Registry Study. Anesth Analg 2024; 139:997-1005. [PMID: 39058628 DOI: 10.1213/ane.0000000000007015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
BACKGROUND Long-term pulmonary complications have been reported after a coronavirus disease-2019 (COVID-19). We hypothesized that a history of COVID-19 is associated with a measurable decrease in baseline respiratory system compliance in patients undergoing general anesthesia. METHODS In this hospital registry study, we included adult patients undergoing general anesthesia between January 2020 and March 2022 at a tertiary health care network in Massachusetts. We excluded patients with an American Society of Anesthesiologists physical status >IV, laryngoscopic surgeries, and patients who arrived intubated. The primary exposure was a history of COVID-19. The primary outcome was baseline respiratory system compliance (mL/cmH 2 O). Effects of severity of infection, surges (Alpha 1 , Alpha 2 , Delta, and Omicron), patient demographics, and time between infection and assessment of compliance were investigated. RESULTS A total of 19,921 patients were included. Approximately 1386 (7.0%) patients had a history of COVID-19. A history of COVID-19 at any time before surgery was associated with a measurably lower baseline respiratory system compliance (ratio of means adj = 0.96; 95% confidence interval [CI], 0.94-0.97; P < .001; adjusted compliance difference: -1.6 mL/cmH 2 O). The association was more pronounced in patients with a severe form of COVID-19 (ratio of means adj = 0.95; 95% CI, 0.90-0.99; P = .02, adjusted compliance difference: -2 mL/cmH 2 O). Alpha 1 , Alpha 2 , and Delta surges, but not Omicron, led to a lower baseline respiratory system compliance ( P < .001, P = .02, and P < .001). The Delta surge effect was magnified in Hispanic ethnicity ( P -for-interaction = 0.003; ratio of means adj = 0.83; 95% CI, 0.74-0.93; P = .001; adjusted compliance difference: -4.6 mL/cmH 2 O). CONCLUSIONS A history of COVID-19 infection during Alpha 1 , Alpha 2 , and Delta surges was associated with a measurably lower baseline respiratory system compliance.
Collapse
Affiliation(s)
- Aiman Suleiman
- From the Center for Anesthesia Research Excellence (CARE), Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Anesthesia and Intensive Care, Faculty of Medicine, University of Jordan, Amman, Jordan
| | - Ricardo Munoz-Acuna
- From the Center for Anesthesia Research Excellence (CARE), Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Simone Redaelli
- From the Center for Anesthesia Research Excellence (CARE), Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Anesthesia, School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Elena Ahrens
- From the Center for Anesthesia Research Excellence (CARE), Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Tim M Tartler
- From the Center for Anesthesia Research Excellence (CARE), Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Sarah Ashrafian
- From the Center for Anesthesia Research Excellence (CARE), Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - May M Hashish
- From the Center for Anesthesia Research Excellence (CARE), Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Abeer Santarisi
- From the Center for Anesthesia Research Excellence (CARE), Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Guanqing Chen
- From the Center for Anesthesia Research Excellence (CARE), Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Stefan Riedel
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Daniel Talmor
- From the Center for Anesthesia Research Excellence (CARE), Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Elias N Baedorf Kassis
- From the Center for Anesthesia Research Excellence (CARE), Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Pulmonary, Critical Care & Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Maximilian S Schaefer
- From the Center for Anesthesia Research Excellence (CARE), Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Anesthesiology, Universitätsklinikum Düsseldorf, Düsseldorf, Germany
| | - Valerie Goodspeed
- From the Center for Anesthesia Research Excellence (CARE), Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| |
Collapse
|
10
|
Zheng Q, Shen Q, Shu Z, Chang K, Zhong K, Yan Y, Ke J, Huang J, Su R, Xia J, Zhou X. Deep representation learning from electronic medical records identifies distinct symptom based subtypes and progression patterns for COVID-19 prognosis. Int J Med Inform 2024; 191:105555. [PMID: 39089210 DOI: 10.1016/j.ijmedinf.2024.105555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/17/2024] [Accepted: 07/14/2024] [Indexed: 08/03/2024]
Abstract
OBJECTIVE Symptoms are significant kind of phenotypes for managing and controlling of the burst of acute infectious diseases, such as COVID-19. Although patterns of symptom clusters and time series have been considered the high potential prediction factors for the prognosis of patients, the elaborated subtypes and their progression patterns based on symptom phenotypes related to the prognosis of COVID-19 patients still need be detected. This study aims to investigate patient subtypes and their progression patterns with distinct features of outcome and prognosis. METHODS This study included a total of 14,139 longitudinal electronic medical records (EMRs) obtained from four hospitals in Hubei Province, China, involving 2,683 individuals in the early stage of COVID-19 pandemic. A deep representation learning model was developed to help acquire the symptom profiles of patients. K-means clustering algorithm is used to divide them into distinct subtypes. Subsequently, symptom progression patterns were identified by considering the subtypes associated with patients upon admission and discharge. Furthermore, we used Fisher's test to identify significant clinical entities for each subtype. RESULTS Three distinct patient subtypes exhibiting specific symptoms and prognosis have been identified. Particularly, Subtype 0 includes 44.2% of the whole and is characterized by poor appetite, fatigue and sleep disorders; Subtype 1 includes 25.6% cases and is characterized by confusion, cough with bloody sputum, encopresis and urinary incontinence; Subtype 2 includes 30.2% cases and is characterized by dry cough and rhinorrhea. These three subtypes demonstrate significant disparities in prognosis, with the mortality rates of 4.72%, 8.59%, and 0.25% respectively. Furthermore, symptom cluster progression patterns showed that patients with Subtype 0 who manifest dark yellow urine, chest pain, etc. in the admission stage exhibit an elevated risk of transforming into the more severe subtypes with poor outcome, whereas those presenting with nausea and vomiting tend to incline towards entering the milder subtype. CONCLUSION This study has proposed a clinical meaningful approach by utilizing the deep representation learning and real-world EMR data containing symptom phenotypes to identify the COVID-19 subtypes and their progression patterns. The results would be potentially useful to help improve the precise stratification and management of acute infectious diseases.
Collapse
Affiliation(s)
- Qiguang Zheng
- School of Computer and Information Technology, Beijing Jiaotong University, China
| | - Qifan Shen
- School of Computer and Information Technology, Beijing Jiaotong University, China
| | - Zixin Shu
- School of Computer and Information Technology, Beijing Jiaotong University, China
| | - Kai Chang
- School of Computer and Information Technology, Beijing Jiaotong University, China
| | - Kunyu Zhong
- School of Computer and Information Technology, Beijing Jiaotong University, China
| | - Yuhang Yan
- School of Computer and Information Technology, Beijing Jiaotong University, China
| | - Jia Ke
- Hubei Provincial Hospital of Traditional Chinese Medicine, China
| | - Jingjing Huang
- Hubei Provincial Hospital of Traditional Chinese Medicine, China
| | - Rui Su
- Beijing Hospital of Traditional Chinese Medicine, China
| | - Jianan Xia
- School of Computer and Information Technology, Beijing Jiaotong University, China.
| | - Xuezhong Zhou
- School of Computer and Information Technology, Beijing Jiaotong University, China.
| |
Collapse
|
11
|
Rajan MV, Sharma V, Upadhyay N, Murali A, Bandyopadhyay S, Hariprasad G. Serum proteomics for the identification of biomarkers to flag predilection of COVID19 patients to various organ morbidities. Clin Proteomics 2024; 21:61. [PMID: 39487396 PMCID: PMC11531188 DOI: 10.1186/s12014-024-09512-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 10/23/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND COVID19 is a pandemic that has affected millions around the world since March 2020. While many patients recovered completely with mild illness, many patients succumbed to various organ morbidities. This heterogeneity in the clinical presentation of COVID19 infection has posed a challenge to clinicians around the world. It is therefore crucial to identify specific organ-related morbidity for effective treatment and better patient outcomes. We have carried out serum-based proteomic experiments to identify protein biomarkers that can flag organ dysfunctions in COVID19 patients. METHODS COVID19 patients were screened and tested at various hospitals across New Delhi, India. 114 serum samples from these patients, with and without organ morbidities were collected and annotated based on clinical presentation and treatment history. Of these, 29 samples comprising of heart, lung, kidney, gastrointestinal, liver, and neurological morbidities were considered for the discovery phase of the experiment. Proteins were isolated, quantified, trypsin digested, and the peptides were subjected to liquid chromatography assisted tandem mass spectrometry analysis. Data analysis was carried out using Proteome Discoverer software. Fold change analysis was carried out on MetaboAnalyst. KEGG, Reactome, and Wiki Pathway analysis of differentially expressed proteins were carried out using the STRING database. Potential biomarker candidates for various organ morbidities were validated using ELISA. RESULTS 254 unique proteins were identified from all the samples with a subset of 12-31 differentially expressed proteins in each of the clinical phenotypes. These proteins establish complement and coagulation cascade pathways in the pathogenesis of the organ morbidities. Validation experiments along with their diagnostic parameters confirm Secreted Protein Acidic and Rich in Cysteine, Cystatin C, and Catalase as potential biomarker candidates that can flag cardiovascular disease, renal disease, and respiratory disease, respectively. CONCLUSIONS Label free serum proteomics shows differential protein expression in COVID19 patients with morbidity as compared to those without morbidity. Identified biomarker candidates hold promise to flag organ morbidities in COVID19 for efficient patient care.
Collapse
Affiliation(s)
- Madhan Vishal Rajan
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Vipra Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Neelam Upadhyay
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Ananya Murali
- Subbaiah Institute of Medical Sciences, New Delhi, Karnataka, India
| | - Sabyasachi Bandyopadhyay
- Proteomics Sub-Facility, Centralized Core Research Facility, All India Institute of Medical Sciences, New Delhi, India
| | - Gururao Hariprasad
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
12
|
Ma Y, Zhu T, Yang P, Gao R, Shen L, Gao P, Gao J, Liu X, Jiang H, Zhang X. The neurological decline and psychological factors caused by coronavirus disease 2019 may be predictors of erectile dysfunction. Andrology 2024; 12:1851-1856. [PMID: 38436126 DOI: 10.1111/andr.13613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 01/14/2024] [Accepted: 01/24/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND Since the outbreak of coronavirus disease 2019, it has had a serious impact on people's physical and mental health. However, in our clinical work, we have found that the erectile function of coronavirus disease 2019 patients with neurological decline was often seriously affected. OBJECTIVES To further explore the relationship between erectile dysfunction and neurological dysfunction caused by coronavirus disease 2019. MATERIALS AND METHODS We conducted a survey from August 2022 to February 2023 at the First Affiliated Hospital of Anhui Medical University and the Third People's Hospital of Linyi City. A total of 251 subjects with a history of coronavirus disease 2019 infection were included. Symptoms and changes in erectile function after the coronavirus disease 2019 infection were collected and assessed using the International Index of Erectile Function-5 scale and several targeted questions. RESULTS In this study, we found that in patients infected with novel coronavirus, the proportion of erectile dysfunction was higher in those with neurological manifestations such as olfactory and taste impairment or psychological symptoms such as anxiety. DISCUSSION We found that neurological decline and psychological factors were independent and significant risk factors for erectile dysfunction caused by coronavirus disease 2019. CONCLUSION Patients with neurological damage or psychiatric symptoms are more likely to have erectile dysfunction, suggesting that the 2019 novel coronavirus may affect erectile function by damaging nerves. This provides a new insight into the mechanism of erectile dysfunction.
Collapse
Affiliation(s)
- Yukuai Ma
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Tianle Zhu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Peng Yang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Rui Gao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lanhui Shen
- Department of Urology, the Third People's Hospital of Linyi, Linyi, China
| | - Pan Gao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jingjing Gao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xi Liu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hui Jiang
- Department of Urology, Peking University First Hospital Institute of Urology, Peking University Andrology Center, Peking University First Hospital, Hefei, China
| | - Xiansheng Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
13
|
Kraemer S, Schneider DJ, Paterson C, Perry D, Westacott MJ, Hagar Y, Katilius E, Lynch S, Russell TM, Johnson T, Astling DP, DeLisle RK, Cleveland J, Gold L, Drolet DW, Janjic N. Crossing the Halfway Point: Aptamer-Based, Highly Multiplexed Assay for the Assessment of the Proteome. J Proteome Res 2024; 23:4771-4788. [PMID: 39038188 PMCID: PMC11536431 DOI: 10.1021/acs.jproteome.4c00411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/24/2024]
Abstract
Measuring responses in the proteome to various perturbations improves our understanding of biological systems. The value of information gained from such studies is directly proportional to the number of proteins measured. To overcome technical challenges associated with highly multiplexed measurements, we developed an affinity reagent-based method that uses aptamers with protein-like side chains along with an assay that takes advantage of their unique properties. As hybrid affinity reagents, modified aptamers are fully comparable to antibodies in terms of binding characteristics toward proteins, including epitope size, shape complementarity, affinity and specificity. Our assay combines these intrinsic binding properties with serial kinetic proofreading steps to allow highly effective partitioning of stable specific complexes from unstable nonspecific complexes. The use of these orthogonal methods to enhance specificity effectively overcomes the severe limitation to multiplexing inherent to the use of sandwich-based methods. Our assay currently measures half of the unique proteins encoded in the human genome with femtomolar sensitivity, broad dynamic range and exceptionally high reproducibility. Using machine learning to identify patterns of change, we have developed tests based on measurement of multiple proteins predictive of current health states and future disease risk to guide a holistic approach to precision medicine.
Collapse
Affiliation(s)
- Stephan Kraemer
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Daniel J. Schneider
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Clare Paterson
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Darryl Perry
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Matthew J. Westacott
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Yolanda Hagar
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Evaldas Katilius
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Sean Lynch
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Theresa M. Russell
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Ted Johnson
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - David P. Astling
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Robert Kirk DeLisle
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Jason Cleveland
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Larry Gold
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Daniel W. Drolet
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| | - Nebojsa Janjic
- SomaLogic, 2495 Wilderness Place, Boulder, Colorado 80301, United States of America
| |
Collapse
|
14
|
Lund K, Nielsen J, Kjeldsen S, Póvoa P, Knudsen T, Nørgård BM, Kjeldsen J. The Impact of Comorbidity and Age on the Risk of Hospitalization and Mortality in Patients with Previous COVID-19 Infection-Based on Nationwide Data. J Clin Med 2024; 13:6522. [PMID: 39518661 PMCID: PMC11546196 DOI: 10.3390/jcm13216522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Objectives: The influence of comorbidity on long-term hospitalization and mortality after COVID-19 in adults (40-59 years) and older adults (≥60 years) is yet to be explored. Methods: This is a Danish population-based cohort study of patients with a first-time positive PCR test for COVID-19 from 1 March 2020, to 28 February 2022 (N = 1,034,103). Exposed cohorts were patients with 1) a Charlson Comorbidity Index (CCI) score of 1-2 and 2) a CCI score ≥3, who were compared to patients without comorbidity (CCI of zero) within the groups of adults (67.9%) and older adults (32.1%) for the risk of hospitalization and mortality. Next, within the age groups, each disease category of the CCI was considered as an exposed cohort and compared to patients who did not have the specific disease of interest. Adjusted hazard ratios (HR) for hospitalization and mortality were estimated by Cox regression models adjusted for confounders. Results: The highest HRs were in adult patients with a CCI score of ≥3. The adjusted HR was 4.54 (95%CI: 4.38-4.70) for hospitalization, and among older adults it was 3.05 (95%CI: 2.99-3.11). The adjusted HR for mortality among adults with a CCI score ≥3 was 21.04 (95%CI: 18.86-23.47), and the adjusted HR for mortality among older adults was 4.61 (95%CI: 4.44-4.78). The underlying disease influenced the risk estimates among adults and older adults, and "dementia" had the highest impact on mortality. Conclusion: A CCI score of 1 or above increases the risk of hospitalization and mortality up to 2 years after a positive PCR test of COVID-19 for adults and older adults. Further, the type of underlying disease in older adults highly influences the risk of hospitalization and mortality.
Collapse
Affiliation(s)
- Ken Lund
- Center for Clinical Epidemiology, Odense University Hospital, 5000 Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark;
| | - Jan Nielsen
- Center for Clinical Epidemiology, Odense University Hospital, 5000 Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark;
| | - Simon Kjeldsen
- Department of Paediatrics and Adolescent Medicine, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Pedro Póvoa
- Center for Clinical Epidemiology, Odense University Hospital, 5000 Odense, Denmark
- Intensive Care Unit 4, Department of Intensive Care, Hospital de São Francisco Xavier, ULSLO, Estrada do Forte do Alto do Duque, 1449-005 Lisbon, Portugal
- NOVA Medical School, CHRC, New University of Lisbon, Campo dos Mártires da Pátria, 1169-056 Lisbon, Portugal
| | - Torben Knudsen
- Department of Medicine, Hospital of Southwest Jutland, 6700 Esbjerg, Denmark
- Department of Regional Health Science, University of Southern Denmark, 5230 Odense, Denmark
| | - Bente Mertz Nørgård
- Center for Clinical Epidemiology, Odense University Hospital, 5000 Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark;
| | - Jens Kjeldsen
- Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark;
- Department of Medical Gastrointestinal Diseases, Odense University Hospital, 5000 Odense, Denmark
| |
Collapse
|
15
|
Ren Z, Zeng X, Lao Y, Zheng H, You Z, Xiang H, Zou Q. A spatial hierarchical network learning framework for drug repositioning allowing interpretation from macro to micro scale. Commun Biol 2024; 7:1413. [PMID: 39478146 PMCID: PMC11525566 DOI: 10.1038/s42003-024-07107-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/21/2024] [Indexed: 11/02/2024] Open
Abstract
Biomedical network learning offers fresh prospects for expediting drug repositioning. However, traditional network architectures struggle to quantify the relationship between micro-scale drug spatial structures and corresponding macro-scale biomedical networks, limiting their ability to capture key pharmacological properties and complex biomedical information crucial for drug screening and therapeutic discovery. Moreover, challenges such as difficulty in capturing long-range dependencies hinder current network-based approaches. To address these limitations, we introduce the Spatial Hierarchical Network, modeling molecular 3D structures and biological associations into a unified network. We propose an end-to-end framework, SpHN-VDA, integrating spatial hierarchical information through triple attention mechanisms to enhance machine understanding of molecular functionality and improve the accuracy of virus-drug association identification. SpHN-VDA outperforms leading models across three datasets, particularly excelling in out-of-distribution and cold-start scenarios. It also exhibits enhanced robustness against data perturbation, ranging from 20% to 40%. It accurately identifies critical motifs for binding sites, even without protein residue annotations. Leveraging reliability of SpHN-VDA, we have identified 25 potential candidate drugs through gene expression analysis and CMap. Molecular docking experiments with the SARS-CoV-2 spike protein further corroborate the predictions. This research highlights the broad potential of SpHN-VDA to enhance drug repositioning and identify effective treatments for various diseases.
Collapse
Affiliation(s)
- Zhonghao Ren
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Xiangxiang Zeng
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Yizhen Lao
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Heping Zheng
- College of Biology, Department of Molecular Medicine, Hunan University, Changsha, China
| | - Zhuhong You
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Hongxin Xiang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
16
|
Wendt R, Macholz M, Kalbitz S, Herrmann N, Herbst V, Hammes T, Kai M, Ankersmit HJ, Beige J, Lübbert C, Graf A, Scherberich J. Serum uromodulin associates with kidney function and outcome in a cohort of hospitalised COVID-19 patients. Sci Rep 2024; 14:25420. [PMID: 39455668 PMCID: PMC11512065 DOI: 10.1038/s41598-024-76372-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
This study investigates the prevalence and evaluates the prognostic implications of acute kidney injury (AKI) in COVID-19 patients, with a novel emphasis on the evaluation of serum uromodulin (sUmod) as a potential kidney-specific biomarker. A cohort of hospitalised COVID-19 patients (n = 378) was examined for AKI using standard criteria. In addition to traditional urinary biomarkers, sUmod levels were analysed. Univariable and multivariable regression models were employed to evaluate the association of sUmod and AKI and in-hospital mortality. Levels of sUmod were significantly lower in patients with CKD (91.8 ± 60.7 ng/ml) compared to patients with normal kidney function (204.7 ± 91.7 ng/ml; p < 0.001). 151 patients (40.0%) presented with AKI at the time of hospital admission or developed an AKI during hospitalization. 116 patients (76.8%) had an AKI already at the time of hospital admission. COVID-19 patients with AKI had significantly lower levels of sUmod compared to patients without AKI during hospitalisation (124.8 ± 79.5 ng/ml) vs 214.6 ± 92.3 ng/ml; p < 0.001). The in-hospital mortality rate in this cohort of COVID-19 patients was 15.3%. Patients with AKI had a higher probability for in-hospital death (OR 5.6, CI 1.76 to 17.881, p = 0.004). Patients who died during hospital stay, had significantly lower sUmod levels (129.14 ± 89.56 ng/ml) compared to patients surviving hospitalisation (187.71 ± 96,64 ng/ml; p < 0.001). AKI is frequently associated with COVID-19 in hospitalized patients. Serum uromodulin may emerge as a promising biomarker for AKI in COVID-19 patients. Further research is warranted to explore its clinical application and refine risk stratification in this patient population.
Collapse
Affiliation(s)
- Ralph Wendt
- Department of Nephrology, St. Georg Hospital, Delitzscher Str. 141, 04129, Leipzig, Germany.
| | - Martin Macholz
- Department of Infectious Diseases and Tropical Medicine, St. Georg Hospital, Delitzscher Str. 141, 04129, Leipzig, Germany
| | - Sven Kalbitz
- Department of Infectious Diseases and Tropical Medicine, St. Georg Hospital, Delitzscher Str. 141, 04129, Leipzig, Germany
| | - Nadja Herrmann
- Department of Finance and Controlling, St. Georg Hospital, Delitzscher Str. 141, 04129, Leipzig, Germany
| | - Victor Herbst
- Institute for Experimental Immunology, Affiliated to EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560, Lübeck, Germany
| | - Tabea Hammes
- Institute for Experimental Immunology, Affiliated to EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560, Lübeck, Germany
| | - Marco Kai
- Institute for Experimental Immunology, Affiliated to EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560, Lübeck, Germany
| | - Hendrik Jan Ankersmit
- Clinic of Thoracic Surgery, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Joachim Beige
- Kuratorium for Dialysis and Transplantation (KfH) Renal Unit, Hospital St. Georg, Delitzscher Str. 141, 04129, Leipzig, Germany
- Martin-Luther University Halle/Wittenberg, Halle, Germany
| | - Christoph Lübbert
- Department of Infectious Diseases and Tropical Medicine, St. Georg Hospital, Delitzscher Str. 141, 04129, Leipzig, Germany
- Division of Infectious Diseases and Tropical Medicine, Leipzig University Hospital, Liebigstr. 20, 04103, Leipzig, Germany
| | - Alexandra Graf
- Institute of Medical Statistics, Center for Medical Data Science, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Jürgen Scherberich
- Department of Nephrology and Clinical Immunology, Klinikum München, 81545, Munich, Germany
| |
Collapse
|
17
|
Cahuapaza-Gutierrez NL. Aplastic Anemia in the light of the COVID-19 pandemic: infection, vaccination, and pathophysiologic mechanisms. Ann Hematol 2024:10.1007/s00277-024-06052-9. [PMID: 39441353 DOI: 10.1007/s00277-024-06052-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Patients infected with SARS-CoV-2 and vaccinated against COVID-19 could develop aplastic anemia (AA). Comprehensive review and analysis were conducted through a selective literature search in PubMed, Scopus, EMBASE, and Web of Science. For this analysis, 26 studies were included, comprising 16 case reports, 7 case series, and 3 observational studies, totaling 53 patients. The causes of acquired or idiopathic AA are diverse; this review presents recent findings, including possible new etiologies such as SARS-CoV-2 infection and COVID-19 vaccines. This possible association is explored, addressing the existing gap, and aiming to improve daily medical practice. This article reviews the relationship between AA and SARS-CoV-2 infection, as well as COVID-19 vaccines, analyzing cases of de novo occurrence and relapses of AA. Although a definitive mechanistic link has not yet been established, possible underlying pathophysiological mechanisms are explored.
Collapse
Affiliation(s)
- Nelson Luis Cahuapaza-Gutierrez
- Facultad de Ciencias de La Salud, Carrera de Medicina Humana, Universidad Científica del Sur, Lima, Perú.
- CHANGE Research Working Group, Universidad Científica del Sur, Lima, Perú.
| |
Collapse
|
18
|
O'Neil ST, Madlock-Brown C, Wilkins KJ, McGrath BM, Davis HE, Assaf GS, Wei H, Zareie P, French ET, Loomba J, McMurry JA, Zhou A, Chute CG, Moffitt RA, Pfaff ER, Yoo YJ, Leese P, Chew RF, Lieberman M, Haendel MA. Finding Long-COVID: temporal topic modeling of electronic health records from the N3C and RECOVER programs. NPJ Digit Med 2024; 7:296. [PMID: 39433942 PMCID: PMC11494196 DOI: 10.1038/s41746-024-01286-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 10/07/2024] [Indexed: 10/23/2024] Open
Abstract
Post-Acute Sequelae of SARS-CoV-2 infection (PASC), also known as Long-COVID, encompasses a variety of complex and varied outcomes following COVID-19 infection that are still poorly understood. We clustered over 600 million condition diagnoses from 14 million patients available through the National COVID Cohort Collaborative (N3C), generating hundreds of highly detailed clinical phenotypes. Assessing patient clinical trajectories using these clusters allowed us to identify individual conditions and phenotypes strongly increased after acute infection. We found many conditions increased in COVID-19 patients compared to controls, and using a novel method to associate patients with clusters over time, we additionally found phenotypes specific to patient sex, age, wave of infection, and PASC diagnosis status. While many of these results reflect known PASC symptoms, the resolution provided by this unprecedented data scale suggests avenues for improved diagnostics and mechanistic understanding of this multifaceted disease.
Collapse
Affiliation(s)
- Shawn T O'Neil
- Department of Genetics, UNC School of Medicine, Chapel Hill, NC, USA.
| | - Charisse Madlock-Brown
- Health Informatics and Information Management Program, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Kenneth J Wilkins
- Biostatistics Program, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Hannah E Davis
- Patient-Led Research Collaborative (PLRC), Washington, DC, USA
| | - Gina S Assaf
- Patient-Led Research Collaborative (PLRC), Washington, DC, USA
| | - Hannah Wei
- Patient-Led Research Collaborative (PLRC), Washington, DC, USA
| | - Parya Zareie
- University of California Davis Health, Davis, CA, USA
| | - Evan T French
- Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA, USA
| | - Johanna Loomba
- The Integrated Translational Health Research Institute of Virginia (iTHRIV), University of Virginia, Charlottesville, VA, USA
| | - Julie A McMurry
- Department of Genetics, UNC School of Medicine, Chapel Hill, NC, USA
| | - Andrea Zhou
- The Integrated Translational Health Research Institute of Virginia (iTHRIV), University of Virginia, Charlottesville, VA, USA
| | - Christopher G Chute
- Schools of Medicine, Public Health and Nursing, Johns Hopkins University, Baltimore, MD, USA
| | - Richard A Moffitt
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Emily R Pfaff
- NC TraCS Institute, UNC School of Medicine, Chapel Hill, NC, USA
| | - Yun Jae Yoo
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Peter Leese
- NC TraCS Institute, UNC School of Medicine, Chapel Hill, NC, USA
| | - Robert F Chew
- Center for Data Science and AI, RTI International, Research Triangle Park, Durham, NC, USA
| | - Michael Lieberman
- OCHIN, Inc, Portland, OR, USA
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health and Science University, Portland, OR, USA
| | - Melissa A Haendel
- Department of Genetics, UNC School of Medicine, Chapel Hill, NC, USA
| |
Collapse
|
19
|
Betts C, Ahlfinger Z, Udeh MC, Kirmani BF. Recent Updates on COVID-19 Associated Strokes. Neurosci Insights 2024; 19:26331055241287730. [PMID: 39391859 PMCID: PMC11465292 DOI: 10.1177/26331055241287730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024] Open
Abstract
The SARS-CoV-2 virus is primarily a respiratory virus, but, as it spread worldwide, it became apparent that there are multiple extrapulmonary manifestations. Reports arose of young and otherwise healthy patients presenting to emergency departments with large-vessel occlusions. Because of a rapidly evolving pandemic, conflicting data sometimes arose regarding the impact of the pandemic on strokes. COVID-19 can induce a hypercoagulable and a proinflammatory state through the interactions with the ACE-2 receptor. These mechanisms may lead to the strokes, both ischemic and hemorrhagic, that are seen in this infection. Strokes, in conjunction with COVID-19 infection, tended to be more disabling and portended a higher mortality. Treatment of these strokes was challenging, as emergency departments were strained with the high burden of COVID-19 admissions. Finally, vaccines against COVID-19 were widely administered, and their potential to cause stroke as an adverse event are discussed. This article will provide an in depth review of the recent updates about the incidence, epidemiology, pathophysiology, clinical presentation and treatment of strokes that are associated with COVID-19.
Collapse
Affiliation(s)
- Colton Betts
- School of Medicine, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Zane Ahlfinger
- School of Medicine, Texas A&M University Health Science Center, Bryan, TX, USA
| | | | - Batool F Kirmani
- School of Medicine, Texas A&M University Health Science Center, Bryan, TX, USA
- Department of Neurology, CHI St. Joseph Health, Bryan, TX, USA
| |
Collapse
|
20
|
Ammar MM, Ben Said NM, Ben Said YN, Abdelsalam AM, Levushkin SP, Laptev A, Inoubli M, Chlif M. Comparative Analysis of Heart Rate Variability and Arterial Stiffness in Elite Male Athletes after COVID-19. J Clin Med 2024; 13:5990. [PMID: 39408050 PMCID: PMC11477989 DOI: 10.3390/jcm13195990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 09/20/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
This study investigated the long-term cardiovascular effects of coronavirus disease (COVID-19) in elite male athletes by comparing the heart rate variability (HRV), arterial stiffness, and other cardiovascular parameters between those with and without prior COVID-19 infection. Methods: This cross-sectional study evaluated 120 elite male athletes (60 post COVID-19, 60 controls) using anthropometric measurements, body composition analysis, pulmonary function tests, HRV analysis, arterial stiffness assessments, hemodynamic monitoring, and microcirculatory function tests. Results: Athletes post COVID-19 showed significantly higher lean mass (p = 0.007), forced vital capacity (p = 0.001), and forced expiratory volume in 1 s (p = 0.007) than controls. HRV parameters did not significantly differ between the groups. Post-COVID-19 athletes exhibited peripheral vascular resistance (p = 0.048) and reflection index (p = 0.038). No significant differences were observed in the blood pressure, cardiac output, oxygen saturation, or microcirculatory oxygen absorption. Conclusions: Elite male athletes showed notable cardiovascular resilience after COVID-19, with only minor differences in vascular function. The maintained cardiac autonomic function and improved lung parameters in post-COVID-19 athletes suggests an adaptive response. These findings support the cardiovascular health of elite athletes following COVID-19 but emphasize the importance of continued monitoring.
Collapse
Affiliation(s)
- Mohamed M. Ammar
- Exercise Physiology Department, College of Sport Science and Physical Activities, King Saud University, Riyadh 11362, Saudi Arabia
| | - Noureddine M. Ben Said
- Biomechanics and Motor Behavior Department, College of Sport Science and Physical Activities, King Saud University, Riyadh 12371, Saudi Arabia; (N.M.B.S.); (A.M.A.)
| | | | - Ahmed M. Abdelsalam
- Biomechanics and Motor Behavior Department, College of Sport Science and Physical Activities, King Saud University, Riyadh 12371, Saudi Arabia; (N.M.B.S.); (A.M.A.)
| | - Sergey P. Levushkin
- Research Institute of Sports and Sports Medicine, Russian University of Sports «GTSOLIFK», Moscow 105122, Russia;
| | - Aleksey Laptev
- Laboratory of Scientific and Methodological Support for Athletes of National Teams, Institute of Sports and Sports Medicine, Moscow 105122, Russia;
| | - Mokhtar Inoubli
- Research Laboratory of Exercise Performance, Health, and Society, Institute of Sport and Physical Education, Manouba University, La Manouba 2010, Tunisia;
| | - Mehdi Chlif
- EA 3300, Exercise Physiology and Rehabilitation Laboratory, Sport Sciences Department, Picardie Jules Verne University, F-80025 Amiens, France
- National Center of Medicine and Science in Sports (NCMSS), Tunisian Research Laboratory Sports Performance Optimization, El Menzah, Tunis 263, Tunisia
| |
Collapse
|
21
|
Gallo A, Murace CA, Corbo MM, Sarlo F, De Ninno G, Baroni S, Fancello G, Masucci L, Covino M, Tosato M, Landi F, Montalto M. Intestinal Inflammation and Permeability in Patients Recovered from SARS-CoV-2 Infection. Dig Dis 2024:1-10. [PMID: 39369712 DOI: 10.1159/000540381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 07/01/2024] [Indexed: 10/08/2024]
Abstract
INTRODUCTION Different works suggest a close link between long COVID gastrointestinal (GI) manifestations and the post-infection disorders of gut-brain interaction (PI-DGBIs). However, the actual mechanisms underlying long-term GI sequelae are still not clear. Our study was aimed to assess both intestinal inflammation and permeability among subjects recovered from SARS-CoV-2 infection and their eventual correlation with long-term GI sequelae. METHODS Eighty-six subjects attending the post-COVID service and recovered from SARS-CoV-2 infection for 6 months were investigated for long COVID manifestations. Those subjects complaining of long-term GI symptoms were further evaluated by Rome IV questionnaire to assess PI-DGBIs. Intestinal inflammation (by fecal calprotectin, FC) and permeability (by serum and fecal levels of zonulin) were evaluated in all subjects. The Hospital Anxiety and Depression Scale (HADS) and the Gastrointestinal Quality of Life Index (GIQLI) questionnaires were further provided to all participants. RESULTS Thirty-seven subjects (43%) complained of long-term GI symptoms, while 49 subjects (57%) did not. Thirty-three subjects fulfilled Rome IV criteria for PI-DGBIs. FC values resulted higher in those subjects who did not complain GI symptoms (p = 0.03), although remaining quite close to the normal range. No significant differences were shown regarding the assessment of intestinal permeability. By GIQLI, long-term GI sequelae were inversely correlated with quality of life (p = 0.009). CONCLUSION Long COVID GI complaints unlikely recognize underlying local inflammatory mechanisms. Since the healthcare, economic, and social burden of post-COVID DGBIs, a deeper understanding of this emerging condition should be encouraged to improve management of the affected subjects.
Collapse
Affiliation(s)
- Antonella Gallo
- Department of Geriatrics and Orthopedics, Fondazione Policlinico Universitario "A. Gemelli," IRCCS, Rome, Italy
| | - Celeste Ambra Murace
- Department of Geriatrics and Orthopedics, Catholic University of the Sacred Heart, Rome, Italy,
| | - Michela Maria Corbo
- Department of Geriatrics and Orthopedics, Catholic University of the Sacred Heart, Rome, Italy
| | - Francesca Sarlo
- Department of Chemistry, Biochemistry and Clinical Molecular Biology, Fondazione Policlinico Universitario "A. Gemelli," IRCCS, Rome, Italy
| | - Grazia De Ninno
- Department of Chemistry, Biochemistry and Clinical Molecular Biology, Catholic University of the Sacred Heart, Rome, Italy
| | - Silvia Baroni
- Department of Chemistry, Biochemistry and Clinical Molecular Biology, Fondazione Policlinico Universitario "A. Gemelli," IRCCS, Rome, Italy
- Department of Chemistry, Biochemistry and Clinical Molecular Biology, Catholic University of the Sacred Heart, Rome, Italy
| | - Giovanni Fancello
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics Fondazione Policlinico Universitario "A. Gemelli," IRCCS, Rome, Italy
| | - Luca Masucci
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics Fondazione Policlinico Universitario "A. Gemelli," IRCCS, Rome, Italy
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics Catholic University of the Sacred Heart, Rome, Italy
| | - Marcello Covino
- Department of Emergency Medicine, Fondazione Policlinico Universitario "A. Gemelli," IRCCS, Rome, Italy
- Catholic University of the Sacred Heart, Rome, Italy
| | - Matteo Tosato
- Department of Geriatrics and Orthopedics, Fondazione Policlinico Universitario "A. Gemelli," IRCCS, Rome, Italy
- Catholic University of the Sacred Heart, Rome, Italy
| | - Francesco Landi
- Department of Geriatrics and Orthopedics, Fondazione Policlinico Universitario "A. Gemelli," IRCCS, Rome, Italy
- Catholic University of the Sacred Heart, Rome, Italy
| | - Massimo Montalto
- Department of Geriatrics and Orthopedics, Fondazione Policlinico Universitario "A. Gemelli," IRCCS, Rome, Italy
- Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
22
|
Medeiros RP, Sousa Marinho R, Magalhães I, Ruão M, Cunha M, Leitão E, Costa R, Neto D, Moreira da Silva Á, Sousa Marinho A. Metabolic Profiles of Critically Ill COVID-19 Patients: A Comparative Analysis of Energy Expenditure. Cureus 2024; 16:e70980. [PMID: 39507132 PMCID: PMC11539026 DOI: 10.7759/cureus.70980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction and aims The COVID-19 pandemic has placed a significant burden on healthcare systems worldwide, especially in the management of critically ill patients. Accurate assessment of energy expenditure (EE) is crucial for providing optimal nutritional support and improving clinical outcomes in these patients. Indirect calorimetry (IC) is the preferred method for measuring EE, yet its use in critically ill COVID-19 patients has not been extensively documented. This study aims to evaluate EE in critically ill patients with and without COVID-19 to identify differences that may impact nutritional support strategies. Methods We conducted a retrospective observational study at a large tertiary university hospital, focusing on critically ill patients. The study compared calorimetry data between patients with and without COVID-19. Data collected included resting energy expenditure, energy expenditure per kilogram, and respiratory exchange rate quotient using IC. Statistical analysis was conducted using SPSS Version 28, with a significance level set at p < 0.05. Results The study included 95 critically ill patients, comprising 61 non-COVID-19 patients and 34 COVID-19 patients. It was found that COVID-19 patients had significantly lower Simplified Acute Physiology Score (SAPS) II compared to their non-COVID-19 counterparts. Although no significant differences in energy needs over time were detected between the two groups or within the COVID-19 group alone, there was a trend towards lower energy needs in COVID-19 patients. Significant differences were noted in respiratory quotient among COVID-19 patients, particularly between those with and without pulmonary thromboembolism, with the latter group exhibiting lower RQ values. Conclusion Our study suggests that while overall EE may not significantly differ between critically ill COVID-19 and non-COVID-19 patients, there are potential differences in substrate use that underline the necessity for individualized nutritional support. These findings highlight the importance of tailoring nutritional interventions to meet the specific needs of critically ill patients, particularly those with COVID-19.
Collapse
Affiliation(s)
- Rita P Medeiros
- Intensive Care Unit, Centro Hospitalar Universitário do Porto, Porto, PRT
| | | | - Inês Magalhães
- Intensive Care Unit, Instituto Ciências Biomédicas Abel Salazar, Porto, PRT
| | - Maria Ruão
- Intensive Care Unit, Centro Hospitalar Universitário do Porto, Porto, PRT
| | - Marisa Cunha
- Intensive Care Unit, Centro Hospitalar Universitário de Santo António, Porto, PRT
| | - Eduarda Leitão
- Intensive Care Unit, Centro Hospitalar Universitário do Porto, Porto, PRT
| | - Rita Costa
- Intensive Care Unit, Centro Hospitalar Universitário do Porto, Porto, PRT
| | - Daniela Neto
- Intensive Care Unit, Centro Hospitalar Universitário do Porto, Porto, PRT
| | | | | |
Collapse
|
23
|
Kole C, Stefanou Ε, Karvelas N, Schizas D, Toutouzas KP. Acute and Post-Acute COVID-19 Cardiovascular Complications: A Comprehensive Review. Cardiovasc Drugs Ther 2024; 38:1017-1032. [PMID: 37209261 PMCID: PMC10199303 DOI: 10.1007/s10557-023-07465-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/02/2023] [Indexed: 05/22/2023]
Abstract
PURPOSE OF REVIEW The risk of cardiovascular complications due to SARS-CoV-2 are significantly increased within the first 6 months of the infection. Patients with COVID-19 have an increased risk of death, and there is evidence that many may experience a wide range of post-acute cardiovascular complications. Our work aims to provide an update on current clinical aspects of diagnosis and treatment of cardiovascular manifestations during acute and long-term COVID-19. RECENT FINDINGS SARS-CoV-2 has been shown to be associated with increased incidence of cardiovascular complications such as myocardial injury, heart failure, and dysrhythmias, as well as coagulation abnormalities not only during the acute phase but also beyond the first 30 days of the infection, associated with high mortality and poor outcomes. Cardiovascular complications during long-COVID-19 were found regardless of comorbidities such as age, hypertension, and diabetes; nevertheless, these populations remain at high risk for the worst outcomes during post-acute COVID-19. Emphasis should be given to the management of these patients. Treatment with low-dose oral propranolol, a beta blocker, for heart rate management may be considered, since it was found to significantly attenuate tachycardia and improve symptoms in postural tachycardia syndrome, while for patients on ACE inhibitors or angiotensin-receptor blockers (ARBs), under no circumstances should these medications be withdrawn. In addition, in patients at high risk after hospitalization due to COVID-19, thromboprophylaxis with rivaroxaban 10 mg/day for 35 days improved clinical outcomes compared with no extended thromboprophylaxis. In this work we provide a comprehensive review on acute and post-acute COVID-19 cardiovascular complications, symptomatology, and pathophysiology mechanisms. We also discuss therapeutic strategies for these patients during acute and long-term care and highlight populations at risk. Our findings suggest that older patients with risk factors such as hypertension, diabetes, and medical history of vascular disease have worse outcomes during acute SARS-CoV-2 infection and are more likely to develop cardiovascular complications during long-COVID-19.
Collapse
Affiliation(s)
- Christo Kole
- Cardiology Department, Sismanoglio General Hospital of Attica, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Εleni Stefanou
- Artificial Kidney Unit, General Hospital of Messinia, Kalamata, Greece
| | - Nikolaos Karvelas
- Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Schizas
- First Department of Surgery, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece
| | | |
Collapse
|
24
|
Tan X, Gao X, Zheng H, Yuan H, Liu H, Ran Q, Luo M. Platelet dysfunction caused by differentially expressed genes as key pathogenic mechanisms in COVID-19. Minerva Cardiol Angiol 2024; 72:517-534. [PMID: 38804627 DOI: 10.23736/s2724-5683.24.06501-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
At the end of 2019, the novel coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) became prevalent worldwide, which brought a heavy medical burden and tremendous economic losses to the world population. In addition to the common clinical respiratory symptoms such as fever, cough and headache, patients with COVID-19 often have hematological diseases, especially platelet dysfunction. Platelet dysfunction usually leads to multiple organ dysfunction, which is closely related to patient severity or mortality. In addition, studies have confirmed significant changes in the gene expression profile of circulating platelets under SARS-CoV-2 infection, which will further lead to changes in platelet function. At the same time, studies have shown that platelets may absorb SARS-COV-2 mRNA independently of ACE2, which further emphasizes the importance of the stability of platelet function in defense against SARS-CoV-2 infection. This study reviewed the relationship between COVID-19 and platelet and SARS-CoV-2 damage to the circulatory system, and further analyzed the significantly differentially expressed mRNA in platelets after infection with SARS-CoV-2 on the basis of previous studies. The top eight hub genes were identified as NLRP3, MT-CO1, CD86, ICAM1, MT-CYB, CASP8, CXCL8 and CXCR4. Subsequently, the effects of SARS-CoV-2 infection on platelet transcript abnormalities and platelet dysfunction were further explored on the basis of 8 hub genes. Finally, the treatment measures of complications caused by platelet dysfunction in patients with COVID-19 were discussed in detail, so as to provide reference for the prevention, diagnosis and treatment of COVID-19.
Collapse
Affiliation(s)
- Xiaoyong Tan
- Department of Pharmacy, Xuanhan County People's Hospital, Dazhou, China
| | - Xiaojun Gao
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Luzhou, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Huanhuan Zheng
- School of Public Health, Southwest Medical University, Luzhou, China
| | - Hui Yuan
- Department of Clinical Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Hong Liu
- Department of Pharmacy, Xuanhan County People's Hospital, Dazhou, China
| | - Qijun Ran
- Department of Pharmacy, Xuanhan County People's Hospital, Dazhou, China
| | - Mao Luo
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, China -
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Drug Discovery Research Center, Southwest Medical University, Luzhou, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
25
|
Krishna B, Metaxaki M, Sithole N, Landín P, Martín P, Salinas-Botrán A. Cardiovascular disease and covid-19: A systematic review. IJC HEART & VASCULATURE 2024; 54:101482. [PMID: 39189008 PMCID: PMC11345335 DOI: 10.1016/j.ijcha.2024.101482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/12/2024] [Accepted: 07/29/2024] [Indexed: 08/28/2024]
Abstract
Background Cardiovascular complications of COVID-19 are numerous and aspects of this phenomenon are not well known. The main objective of this manuscript is a systematic review of the acute and chronic cardiovascular complications secondary to COVID-19. Methods A systematic review of the literature through Medline via PubMed was conducted (2020-2024). Results There is a plethora of effects of COVID-19 on the heart in the acute setting. Here we discuss pathophysiology, myocardial infarctions, heart failure, Takotsubo Cardiomyopathy, myocardial injury, myocarditis and arrhythmias that are caused by COVID-19. Additionally, these cardiovascular injuries can linger and may be an underlying cause of some Long COVID symptoms. Conclusions Cardiovascular complications of COVID-19 are numerous and life-threatening. Long COVID can affect cardiovascular health. Microclotting induced by SARS-CoV-2 infection could be a therapeutic target for some aspects of Long Covid.
Collapse
Affiliation(s)
- B.A. Krishna
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - M. Metaxaki
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - N. Sithole
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- Department of Infectious Diseases, Cambridge University, Cambridge, United Kingdom
| | - P. Landín
- Department of Cardiology, Hospital Clínico San Carlos, Madrid, Spain
| | - P. Martín
- Department of Respiratory Medicine, Hospital Clínico San Carlos, Madrid, Spain
| | - A. Salinas-Botrán
- Department of Infectious Diseases, Hospital Clínico San Carlos, Madrid, Spain
- Department of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
26
|
du Preez HN, Lin J, Maguire GEM, Aldous C, Kruger HG. COVID-19 vaccine adverse events: Evaluating the pathophysiology with an emphasis on sulfur metabolism and endotheliopathy. Eur J Clin Invest 2024; 54:e14296. [PMID: 39118373 DOI: 10.1111/eci.14296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024]
Abstract
In this narrative review, we assess the pathophysiology of severe adverse events that presented after vaccination with DNA and mRNA vaccines against COVID-19. The focus is on the perspective of an undersulfated and degraded glycocalyx, considering its impact on immunomodulation, inflammatory responses, coagulation and oxidative stress. The paper explores various factors that lead to glutathione and inorganic sulfate depletion and their subsequent effect on glycocalyx sulfation and other metabolites, including hormones. Components of COVID-19 vaccines, such as DNA and mRNA material, spike protein antigen and lipid nanoparticles, are involved in possible cytotoxic effects. The common thread connecting these adverse events is endotheliopathy or glycocalyx degradation, caused by depleted glutathione and inorganic sulfate levels, shear stress from circulating nanoparticles, aggregation and formation of protein coronas; leading to imbalanced immune responses and chronic release of pro-inflammatory cytokines, ultimately resulting in oxidative stress and systemic inflammatory response syndrome. By understanding the underlying pathophysiology of severe adverse events, better treatment options can be explored.
Collapse
Affiliation(s)
- Heidi N du Preez
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Durban, South Africa
- College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Johnson Lin
- School of Life Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Glenn E M Maguire
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Durban, South Africa
- School of Chemistry and Physics, University of KwaZulu-Natal, Durban, South Africa
| | - Colleen Aldous
- College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Hendrik G Kruger
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
27
|
Vale C, Godolphin PJ, Fisher D, Horby PW, Kosiborod MN, Hochman JS, Webster K, Higgins JPT, Althouse AD, Berwanger O, Furtado RHM, Gasparyan SB, Haynes R, Koch GG, Landray M, Leifer E, Marshall J, Murthy S, Neal MD, Staplin N, Diaz J, Sterne JAC, Shankar-Hari M. Sodium-glucose co-transporter-2 inhibitors for hospitalised patients with COVID-19: a prospective meta-analysis of randomised trials. Lancet Diabetes Endocrinol 2024; 12:735-747. [PMID: 39250923 DOI: 10.1016/s2213-8587(24)00219-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 02/13/2024] [Accepted: 07/12/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND Sodium-glucose co-transporter-2 (SGLT2) inhibitors have been proposed as a potential treatment for adults hospitalised with COVID-19, due to their potential anti-inflammatory and endothelial protective effects. Published evidence from randomised control trials (RCTs) does not provide evidence of benefit. We aimed to estimate the effect of oral administration of SGLT2 inhibitors compared with usual care or placebo in adults hospitalised with COVID-19. METHODS Eligible RCTs that estimated the effect of oral administration of SGLT2 inhibitors compared with usual care or placebo on 28-day all-cause mortality (primary outcome) were included in this prospective meta-analysis. The primary safety outcome was ketoacidosis by 28 days. Trials were identified through systematic searches of ClinicalTrials.gov, EudraCT, and the WHO ISRCTN registry between Nov 1, 2022 and Jan 31, 2023. The search terms were "random*" AND "COVID" AND each SGLT2i, not restricted by trial status or language. Individual searches were then combined. Prespecified summary outcome data, overall and within subgroups of interest, were provided by each trial. The primary analyses were inverse variance weighted meta-analysis of odds ratios (ORs). Risk of bias was assessed using the Cochrane Risk of Bias tool. This study was registered with PROSPERO, CRD42023406442. FINDINGS Three eligible trials randomly assigned 6096 participants (3025 to the SGLT2 inhibitor group and 3071 to the usual care or placebo group). 2381 (39%) patients were women and 1547 (25%) had type 2 diabetes at randomisation. By 28 days, there were 351 deaths in the SGLT2 inhibitor group and 382 deaths in the usual care or placebo group (summary OR 0·93 [95% CI 0·79-1·08]; p=0·33, I2 for inconsistency across trials 0%). The risk of bias was assessed as being low. Ketoacidosis was observed in seven participants in the SGLT2 inhibitor group and two patients in the usual care or placebo group. INTERPRETATION Although administration of SGLT2 inhibitor was safe, we found no clear evidence that adding SGLT2 inhibitor therapy improved outcomes in patients hospitalised with COVID-19 compared with usual care or placebo. These data do not support the use of SGLT2 inhibitors as standard treatment in adults hospitalised for COVID-19. FUNDING None.
Collapse
Affiliation(s)
- Claire Vale
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, UCL, London, UK
| | - Peter J Godolphin
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, UCL, London, UK
| | - David Fisher
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, UCL, London, UK
| | - Peter W Horby
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mikhail N Kosiborod
- Saint Luke's Mid America Heart Institute, and University of Missouri-Kansas City, Kansas City, MO, USA
| | - Judith S Hochman
- New York University Grossman School of Medicine, New York, NY, USA
| | - Katie Webster
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Julian P T Higgins
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | | | - Otavio Berwanger
- George Institute for Global Health UK at Imperial College London, London, UK
| | - Remo H M Furtado
- Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Samvel B Gasparyan
- Late-stage Development, CVRM, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Richard Haynes
- MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Gary G Koch
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Martin Landray
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Eric Leifer
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - John Marshall
- Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Srinivas Murthy
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | | | - Natalie Staplin
- MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Janet Diaz
- Clinical Unit, Health Emergencies Programme, World Health Organization, Geneva, Switzerland
| | - Jonathan A C Sterne
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK; HDR UK South West, Bristol, UK; NIHR Bristol Biomedical Research Centre, Bristol, UK
| | - Manu Shankar-Hari
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
28
|
Jeon D, Kim SH, Kim J, Jeong H, Uhm C, Oh H, Cho K, Cho Y, Park IH, Oh J, Kim JJ, Hwang JY, Lee HJ, Lee HY, Seo JY, Shin JS, Seong JK, Nam KT. Discovery of a new long COVID mouse model via systemic histopathological comparison of SARS-CoV-2 intranasal and inhalation infection. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167347. [PMID: 39019092 DOI: 10.1016/j.bbadis.2024.167347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 07/19/2024]
Abstract
Intranasal infection is commonly used to establish a SARS-CoV-2 mouse model due to its non-invasive procedures and a minimal effect from the operation itself. However, mice intranasally infected with SARS-CoV-2 have a high mortality rate, which limits the utility of this model for exploring therapeutic strategies and the sequelae of non-fatal COVID-19 cases. To resolve these limitations, an aerosolised viral administration method has been suggested. However, an in-depth pathological analysis comparing the two models is lacking. Here, we show that inhalation and intranasal SARS-CoV-2 (106 PFU) infection models established in K18-hACE2 mice develop unique pathological features in both the respiratory and central nervous systems, which could be directly attributed to the infection method. While the inhalation-infection model exhibited relatively milder pathological parameters, it closely mimicked the prevalent chest CT pattern observed in COVID-19 patients with focal, peripheral lesions and fibrotic scarring in the recuperating lung. We also found the evidence of direct neuron-invasion from the olfactory receptor neurons to the olfactory bulb in the intranasal model and showed the trigeminal nerve as an alternative route of transmission to the brain in inhalation infected mice. Even after viral clearance confirmed at 14 days post-infection, mild lesions were still found in the brain of inhalation-infected mice. These findings suggest that the inhalation-infection model has advantages over the intranasal-infection model in closely mimicking the pathological features of non-fatal symptoms of COVID-19, demonstrating its potential to study the sequelae and possible interventions for long COVID.
Collapse
Affiliation(s)
- Donghun Jeon
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung-Hee Kim
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Jiseon Kim
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Haengdueng Jeong
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Chanyang Uhm
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Heeju Oh
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Kyungrae Cho
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Yejin Cho
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - In Ho Park
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea; Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea
| | - Jooyeon Oh
- Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea
| | - Jeong Jin Kim
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Ji-Yeon Hwang
- Preclinical Research Center, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Hyo-Jung Lee
- Department of Periodontology, Section of Dentistry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Ho-Young Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea; Department of Nuclear Medicine, Seoul National University, College of Medicine, Seoul, South Korea
| | - Jun-Young Seo
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Jeon-Soo Shin
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea; Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea; Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea
| | - Je Kyung Seong
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, South Korea; Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, BK 21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, South Korea; BIO MAX Institute, Seoul National University, Seoul, South Korea; Interdisciplinary Program for Bioinformatics, Seoul National University, Seoul, South Korea.
| | - Ki Taek Nam
- Department of Biomedical Sciences, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
29
|
Pfaender S, Steinmann E. Editorial overview: Coronaviruses 2024. Curr Opin Microbiol 2024; 81:102523. [PMID: 39098125 DOI: 10.1016/j.mib.2024.102523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/06/2024]
Affiliation(s)
- Stephanie Pfaender
- Research Unit Emerging Viruses, Leibniz Institute of Virology (LIV), Hamburg, Germany; University of Lübeck, Lübeck, Germany.
| | - Eike Steinmann
- Department of Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany; German Centre for Infection Research (DZIF), External Partner Site, Bochum, Germany.
| |
Collapse
|
30
|
Ariza M, Delas B, Rodriguez B, De Frutos B, Cano N, Segura B, Barrué C, Bejar J, Asaad M, Cortés CU, Junqué C, Garolera M. Retinal Microvasculature Changes Linked to Executive Function Impairment after COVID-19. J Clin Med 2024; 13:5671. [PMID: 39407733 PMCID: PMC11477391 DOI: 10.3390/jcm13195671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/06/2024] [Accepted: 09/21/2024] [Indexed: 10/20/2024] Open
Abstract
Background/Objectives: Studies using optical coherence tomography angiography (OCTA) have revealed that individuals recovering from COVID-19 have a reduced retinal vascular density (VD) and larger foveal avascular zones (FAZs) than healthy individuals, with more severe cases showing greater reductions. We aimed to examine aspects of the retinal microvascularization in patients with post-COVID-19 condition (PCC) classified by COVID-19 severity and how these aspects relate to cognitive performance. Methods: This observational cross-sectional study included 104 PCC participants from the NAUTILUS Project, divided into severe (n = 59) and mild (n = 45) COVID-19 groups. Participants underwent cognitive assessments and OCTA to measure VD and perfusion density (PD) in the superficial capillary plexus (SVP) and FAZ. Analysis of covariance and partial Pearson and Spearman correlations were used to study intergroup differences and the relationships between cognitive and OCTA variables. Results: Severe PCC participants had significantly lower central (p = 0.03) and total (p = 0.03) VD, lower central (p = 0.02) PD measurements, and larger FAZ areas (p = 0.02) and perimeters (p = 0.02) than mild cases. Severe cases showed more cognitive impairment, particularly in speed processing (p = 0.003) and executive functions (p = 0.03). Lower central VD, lower central PD, and larger FAZ areas and perimeters were associated with worse executive function performance in the entire PCC sample and in the mild COVID-19 group. Conclusions: Retinal microvascular alterations, characterized by reduced VD and PD in the SVP and larger FAZ areas, were associated with cognitive impairments in PCC individuals. These findings suggest that severe COVID-19 leads to long-lasting microvascular damage, impacting retinal and cognitive health.
Collapse
Affiliation(s)
- Mar Ariza
- Grup de Recerca en Cervell, Cognició i Conducta, Consorci Sanitari de Terrassa (CST)-Hospital Universitari, 08227 Terrassa, Spain; (M.A.); (N.C.)
- Unitat de Psicologia Mèdica, Departament de Medicina, Universitat de Barcelona (UB), 08036 Barcelona, Spain
| | - Barbara Delas
- Ophtalmology Department, Consorci Sanitari de Terrassa (CST)-Hospital Universitari, 08227 Terrassa, Spain; (B.D.); (B.R.); (B.D.F.)
| | - Beatriz Rodriguez
- Ophtalmology Department, Consorci Sanitari de Terrassa (CST)-Hospital Universitari, 08227 Terrassa, Spain; (B.D.); (B.R.); (B.D.F.)
| | - Beatriz De Frutos
- Ophtalmology Department, Consorci Sanitari de Terrassa (CST)-Hospital Universitari, 08227 Terrassa, Spain; (B.D.); (B.R.); (B.D.F.)
| | - Neus Cano
- Grup de Recerca en Cervell, Cognició i Conducta, Consorci Sanitari de Terrassa (CST)-Hospital Universitari, 08227 Terrassa, Spain; (M.A.); (N.C.)
- Departament de Ciències Bàsiques, Universitat Internacional de Catalunya, 08195 Sant Cugat del Vallès, Spain
| | - Bàrbara Segura
- Unitat de Psicologia Mèdica, Departament de Medicina, Universitat de Barcelona (UB), 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Institut de Neurociències, Universitat de Barcelona (UB), 08035 Barcelona, Spain
| | - Cristian Barrué
- Departament de Ciències de la Computació, Universitat Politècnica de Catalunya-BarcelonaTech, 08034 Barcelona, Spain (C.U.C.)
| | - Javier Bejar
- Departament de Ciències de la Computació, Universitat Politècnica de Catalunya-BarcelonaTech, 08034 Barcelona, Spain (C.U.C.)
| | - Mouafk Asaad
- Ophtalmology Department, Consorci Sanitari de Terrassa (CST)-Hospital Universitari, 08227 Terrassa, Spain; (B.D.); (B.R.); (B.D.F.)
| | - Claudio Ulises Cortés
- Departament de Ciències de la Computació, Universitat Politècnica de Catalunya-BarcelonaTech, 08034 Barcelona, Spain (C.U.C.)
| | - Carme Junqué
- Unitat de Psicologia Mèdica, Departament de Medicina, Universitat de Barcelona (UB), 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Institut de Neurociències, Universitat de Barcelona (UB), 08035 Barcelona, Spain
| | - Maite Garolera
- Grup de Recerca en Cervell, Cognició i Conducta, Consorci Sanitari de Terrassa (CST)-Hospital Universitari, 08227 Terrassa, Spain; (M.A.); (N.C.)
- Departament de Ciències Bàsiques, Universitat Internacional de Catalunya, 08195 Sant Cugat del Vallès, Spain
- Neuropsychology Unit, Consorci Sanitari de Terrassa (CST)-Hospital Universitari, 08227 Terrassa, Spain
| | | |
Collapse
|
31
|
Aljuaid A. Increased Activation Markers of Adaptive Immunity in Patients with Severe COVID-19. J Clin Med 2024; 13:5664. [PMID: 39407725 PMCID: PMC11477269 DOI: 10.3390/jcm13195664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 09/04/2024] [Accepted: 09/18/2024] [Indexed: 10/20/2024] Open
Abstract
Introduction: COVID-19 is a pandemic disease and is widespread over the world. This disease shows a 5.1% mortality. The understanding of the disease has expanded rapidly in many areas, including virological, epidemiological, clinical, and management dimensions. To better understand the inflammatory and immune profiles that impact the pathogenesis and development of severe COVID-19 symptoms, further studies are essential. This research aims to explore the inflammatory and adaptive immune responses associated with COVID-19, considering factors such as genetic diversity and environmental exposure among Saudi patients. The goal is to determine if patients with severe COVID-19 exhibit different disease phenotypes. Materials and Methods: This case-control study includes 115 participants (healthy and with COVID-19 infection), 55 of which had confirmed cases of COVID-19 in intensive care units (ICUs) at different hospitals in Makkah City, Saudi Arabia. Whole blood samples were collected from June to September 2021 for cellular analyses, and inflammation marker data were collected from hospital records. The expression of activation markers on B (CD27 and CD38) and T cells (CD27 and HLA-DR) was obtained using the flow cytometry technique. Also, serum was collected for cytokine measurements, including IL-6, INF-γ, and TNF- α. Results: The results indicated that lymphopenia and excessive T cell activation were more prevalent in severe cases than in healthy individuals. Furthermore, the results revealed that severe COVID-19 patients had an increased frequency of CD19+ B cells, with changes in B cell subsets. The current study implies impairment and changes in the phenotype of adaptive cells (including T and B cells), with an increase in HLA-DR molecules and inflammation markers with pro-inflammatory cytokines in severe COVID-19 cases. Conclusions: The current study implies impairment and changes in the phenotype of adaptive cells (including T and B cells), with an increase in HLA-DR molecules and inflammation markers in severe COVID-19 cases, which could be targeted for therapeutic interventions. This might be a valuable approach for the diagnosis and treatment of severe COVID-19 cases.
Collapse
Affiliation(s)
- Abdulelah Aljuaid
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
32
|
Triantafyllis AS, Sfantou D, Karapedi E, Peteinaki K, Kotoulas SC, Saad R, Fountoulakis PN, Tsamakis K, Tsiptsios D, Rallidis L, Tsoporis JN, Varvarousis D, Hamodraka E, Giannakopoulos A, Poulimenos LE, Ikonomidis I. Coronary Implications of COVID-19. Med Princ Pract 2024:1-12. [PMID: 39307131 DOI: 10.1159/000541553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024] Open
Abstract
Patients with SARS-CoV-2 infection carry an increased risk of cardiovascular disease encompassing various implications, including acute myocardial injury or infarction, myocarditis, heart failure, and arrhythmias. A growing volume of evidence correlates SARS-CoV-2 infection with myocardial injury, exposing patients to higher mortality risk. SARS-CoV-2 attacks the coronary arterial bed with various mechanisms including thrombosis/rupture of preexisting atherosclerotic plaque, de novo coronary thrombosis, endotheliitis, microvascular dysfunction, vasculitis, vasospasm, and ectasia/aneurysm formation. The angiotensin-converting enzyme 2 receptor plays pivotal role on the cardiovascular homeostasis and the unfolding of COVID-19. The activation of immune system, mediated by proinflammatory cytokines along with the dysregulation of the coagulation system, can pose an insult on the coronary artery, which usually manifests as an acute coronary syndrome (ACS). Electrocardiogram, echocardiography, cardiac biomarkers, and coronary angiography are essential tools to set the diagnosis. Revascularization is the first-line treatment in all patients with ACS and obstructed coronary arteries, whereas in type 2 myocardial infarction treatment of hypoxia, anemia and systemic inflammation are indicated. In patients presenting with coronary vasospasm, nitrates and calcium channel blockers are preferred, while treatment of coronary ectasia/aneurysm mandates the use of antiplatelets/anticoagulants, corticosteroids, immunoglobulin, and biologic agents. It is crucial to untangle the exact mechanisms of coronary involvement in COVID-19 in order to ensure timely diagnosis and appropriate treatment. We have reviewed the current literature and provide a detailed overview of the pathophysiology and clinical spectrum associated with coronary implications of SARS-COV-2 infection.
Collapse
Affiliation(s)
| | - Danai Sfantou
- Department of Cardiology, Asklepeion General Hospital, Athens, Greece
| | - Eleni Karapedi
- Department of Cardiology, Asklepeion General Hospital, Athens, Greece
| | | | | | - Richard Saad
- Department of Cardiology, Asklepeion General Hospital, Athens, Greece
| | | | | | - Dimitrios Tsiptsios
- Department of Neurology, School of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Loukianos Rallidis
- Second Department of Cardiology, Attikon University Hospital, Athens, Greece
| | - James N Tsoporis
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | - Ignatios Ikonomidis
- Second Department of Cardiology, Attikon University Hospital, Athens, Greece
| |
Collapse
|
33
|
Khan A, Ling J, Li J. Is Autophagy a Friend or Foe in SARS-CoV-2 Infection? Viruses 2024; 16:1491. [PMID: 39339967 PMCID: PMC11437447 DOI: 10.3390/v16091491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
As obligate parasites, viruses need to hijack resources from infected cells to complete their lifecycle. The interaction between the virus and host determines the viral infection process, including viral propagation and the disease's outcome. Understanding the interaction between the virus and host factors is a basis for unraveling the intricate biological processes in the infected cells and thereby developing more efficient and targeted antivirals. Among the various fundamental virus-host interactions, autophagy plays vital and also complicated roles by directly engaging in the viral lifecycle and functioning as an anti- and/or pro-viral factor. Autophagy thus becomes a promising target against virus infection. Since the COVID-19 pandemic, there has been an accumulation of studies aiming to investigate the roles of autophagy in SARS-CoV-2 infection by using different models and from distinct angles, providing valuable information for systematically and comprehensively dissecting the interplay between autophagy and SARS-CoV-2. In this review, we summarize the advancements in the studies of the interaction between SARS-CoV-2 and autophagy, as well as detailed molecular mechanisms. We also update the current knowledge on the pharmacological strategies used to suppress SARS-CoV-2 replication through remodeling autophagy. These extensive studies on SARS-CoV-2 and autophagy can advance our understanding of virus-autophagy interaction and provide insights into developing efficient antiviral therapeutics by regulating autophagy.
Collapse
Affiliation(s)
- Asifa Khan
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Uppsala University, P.O. Box 582, 751 23 Uppsala, Sweden
- Biochemistry Unit, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Jiaxin Ling
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Uppsala University, P.O. Box 582, 751 23 Uppsala, Sweden
- Zoonosis Science Center, Uppsala University, 751 23 Uppsala, Sweden
| | - Jinlin Li
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Uppsala University, P.O. Box 582, 751 23 Uppsala, Sweden
| |
Collapse
|
34
|
Attiq A, Afzal S, Wahab HA, Ahmad W, Kandeel M, Almofti YA, Alameen AO, Wu YS. Cytokine Storm-Induced Thyroid Dysfunction in COVID-19: Insights into Pathogenesis and Therapeutic Approaches. Drug Des Devel Ther 2024; 18:4215-4240. [PMID: 39319193 PMCID: PMC11421457 DOI: 10.2147/dddt.s475005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024] Open
Abstract
Angiotensin-converting enzyme 2 receptors (ACE2R) are requisite to enter the host cells for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). ACE2R is constitutive and functions as a type I transmembrane metallo-carboxypeptidase in the renin-angiotensin system (RAS). On thyroid follicular cells, ACE2R allows SARS-CoV-2 to invade the thyroid gland, impose cytopathic effects and produce endocrine abnormalities, including stiff back, neck pain, muscle ache, lethargy, and enlarged, inflamed thyroid gland in COVID-19 patients. Further damage is perpetuated by the sudden bursts of pro-inflammatory cytokines, which is suggestive of a life-threatening syndrome known as a "cytokine storm". IL-1β, IL-6, IFN-γ, and TNF-α are identified as the key orchestrators of the cytokine storm. These inflammatory mediators upregulate transcriptional turnover of nuclear factor-kappa B (NF-κB), Janus kinase/signal transducer and activator of transcription (JAK/STAT), and mitogen-activated protein kinase (MAPK), paving the pathway for cytokine storm-induced thyroid dysfunctions including euthyroid sick syndrome, autoimmune thyroid diseases, and thyrotoxicosis in COVID-19 patients. Targeted therapies with corticosteroids (dexamethasone), JAK inhibitor (baricitinib), nucleotide analogue (remdesivir) and N-acetyl-cysteine have demonstrated effectiveness in terms of attenuating the severity and frequency of cytokine storm-induced thyroid dysfunctions, morbidity and mortality in severe COVID-19 patients. Here, we review the pathogenesis of cytokine storms and the mechanisms and pathways that establish the connection between thyroid disorder and COVID-19. Moreover, cross-talk interactions of signalling pathways and therapeutic strategies to address COVID-19-associated thyroid diseases are also discussed herein.
Collapse
Affiliation(s)
- Ali Attiq
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, Penang, 11800, Malaysia
| | - Sheryar Afzal
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al Ahsa, 31982, Saudi Arabia
| | - Habibah A Wahab
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, Penang, 11800, Malaysia
| | - Waqas Ahmad
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, Penang, 11800, Malaysia
| | - Mahmoud Kandeel
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al Ahsa, 31982, Saudi Arabia
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrel Sheikh, 6860404, Egypt
| | - Yassir A Almofti
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al Ahsa, 31982, Saudi Arabia
- Department of Biochemistry, Molecular Biology and Bioinformatics, College of Veterinary Medicine, University of Bahri, Khartoum, 12217, Sudan
| | - Ahmed O Alameen
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al Ahsa, 31982, Saudi Arabia
- Department of Physiology, Faculty of Veterinary Medicine, University of Khartoum, Shambat, 13314, Sudan
| | - Yuan Seng Wu
- Sunway Microbiome Centre, School of Medical and Life Sciences, Sunway University, Subang Jaya, Selangor, 47500, Malaysia
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Subang Jaya, Selangor, 47500, Malaysia
| |
Collapse
|
35
|
Abbasi A, Gattoni C, Iacovino M, Ferguson C, Tosolini J, Singh A, Soe KK, Porszasz J, Lanks C, Rossiter HB, Casaburi R, Stringer WW. A Pilot Study on the Effects of Exercise Training on Cardiorespiratory Performance, Quality of Life, and Immunologic Variables in Long COVID. J Clin Med 2024; 13:5590. [PMID: 39337079 PMCID: PMC11433403 DOI: 10.3390/jcm13185590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Objectives: Fatigue is a prominent feature of long COVID (LC) and may be related to several pathophysiologic mechanisms, including immune hyperstimulation. Aerobic endurance exercise training may be a useful therapy, with appropriate attention to preventing post-exertional malaise. Methods: Fourteen participants completed a pilot study of aerobic exercise training (twenty 1.5 h sessions of over 10 weeks). Cardiorespiratory fitness, 6 min walk distance, quality of life, symptoms, 7-day physical activity, immunophenotype, and inflammatory biomarkers were measured before and after exercise training. Results: The participant characteristics at baseline were as follows: 53.5 ± 11.6 yrs, 53% f, BMI 32.5 ± 8.4, 42% ex-smokers, 15.1 ± 8.8 months since initial COVID-19 infection, low normal pulmonary function testing, V.O2peak 19.3 ± 5.1 mL/kg/min, 87 ± 17% predicted. After exercise training, participants significantly increased their peak work rate (+16 ± 20 W, p = 0.010) and V.O2peak (+1.55 ± 2.4 mL/kg/min, p = 0.030). Patients reported improvements in fatigue severity (-11%), depression (-42%), anxiety (-29%), and dyspnea level (-46%). There were no changes in 6MW distance or physical activity. The circulating number of CD3+, CD4+, CD19+, CD14++CD16, and CD16++CD14+ monocytes and CD56+ cells (assessed with flow cytometry) increased with acute exercise (rest to peak) and was not diminished or augmented by exercise training. Plasma concentrations of TNF-α, IL-6, IL-8, IL-10, INF-γ, and INF-λ were normal at study entry and not affected by training. Conclusions: Aerobic endurance exercise training in individuals with LC delivered beneficial effects on cardiorespiratory fitness, quality of life, anxiety, depression, and fatigue without detrimental effects on immunologic function.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - William W. Stringer
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (A.A.); (C.G.); (M.I.); (C.F.); (J.T.); (A.S.); (J.P.); (C.L.); (H.B.R.); (R.C.)
| |
Collapse
|
36
|
Wylęgała E, Prus-Ludwig A, Mocek P, Tomczyk T, Dugiełło B, Madej A, Orzechowska-Wylęgała B, Wylęgała A. Retinal and Corneal OCT Results of Patients Hospitalized and Treated in the Acute Phase of COVID-19. J Clin Med 2024; 13:5564. [PMID: 39337050 PMCID: PMC11432357 DOI: 10.3390/jcm13185564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/14/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
Objective: This study aimed to assess changes in the morphology of the retina and cornea in patients treated and hospitalized during the acute active phase of SARS-CoV-2 infection. Methods: A total of 24 patients with symptomatic early COVID-19 disease and 38 healthy participants from a control group were enrolled in our study. Among them, 20 received oxygen therapy at flow rates ranging from 1-10 L, while four received high-flow intranasal oxygen therapy (HFNOT). Some patients were treated with other types of therapy, such as Remdesivir, COVID-19 convalescent plasma therapy, or Tocilizumab. In the study, we focused on the analysis of optical coherence tomography (OCT) images of the cornea and retina including corneal thickness, central retinal thickness, retinal nerve fiber layer (RNFL), and optic disc parameters. The measurements were acquired using Spectral-domain OCT REVO FC 130. Results: The analysis did not show significant changes between the examined ophthalmological parameters before and after therapy. Furthermore, there were no detected significant differences between the tested parameters of the retina and cornea in COVID-19-positive patients compared to the control group. Conclusions: No ophthalmological manifestations of COVID-19 disease were observed during the study. Taking into account the results of other publications, the lack of an unambiguous position on this topic requires further research.
Collapse
Affiliation(s)
- Edward Wylęgała
- Chair and Clinical Department of Ophthalmology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-760 Katowice, Poland
- Department of Ophthalmology, Railway Hospital, 40-760 Katowice, Poland
| | | | - Patrycja Mocek
- Ophthalmology Department, I School of Medicine, Katowice, Medical University of Silesia, 40-514 Katowice, Poland
| | - Tomasz Tomczyk
- Ophthalmology Department, I School of Medicine, Katowice, Medical University of Silesia, 40-514 Katowice, Poland
| | - Bogdan Dugiełło
- Chair and Clinical Department of Ophthalmology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-760 Katowice, Poland
- Department of Ophthalmology, Railway Hospital, 40-760 Katowice, Poland
| | - Andrzej Madej
- Department of Pharmacology, WSB University, 41-300 Dąbrowa Górnicza, Poland
| | - Bogusława Orzechowska-Wylęgała
- Department of Pediatric Otolaryngology, Head and Neck Surgery, Department of Pediatric Surgery, Medical University of Silesia, 40-760 Katowice, Poland
| | - Adam Wylęgała
- Health Promotion and Obesity Management Unit, Department of Pathophysiology, Faculty of Medical Sciences, Medical University of Silesia, 40-752 Katowice, Poland
| |
Collapse
|
37
|
Hou M, Wu J, Li J, Zhang M, Yin H, Chen J, Jin Z, Dong R. Immunothrombosis: A bibliometric analysis from 2003 to 2023. Medicine (Baltimore) 2024; 103:e39566. [PMID: 39287275 PMCID: PMC11404911 DOI: 10.1097/md.0000000000039566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Immunothrombosis is a physiological process that constitutes an intravascular innate immune response. Abnormal immunothrombosis can lead to thrombotic disorders. With the outbreak of COVID-19, there is increasing attention to the mechanisms of immunothrombosis and its critical role in thrombotic events, and a growing number of relevant research papers are emerging. This article employs bibliometrics to discuss the current status, hotspots, and trends in research of this field. METHODS Research papers relevant to immunothrombosis published from January 1, 2003, to May 29, 2023, were collected from the Web of Science Core Collection database. VOSviewer and the R package "Bibliometrix" were employed to analyze publication metrics, including the number of publications, authors, countries, institutions, journals, and keywords. The analysis generated visual results, and trends in research topics and hotspots were examined. RESULTS A total of 495 target papers were identified, originating from 58 countries and involving 3287 authors from 1011 research institutions. Eighty high-frequency keywords were classified into 5 clusters. The current key research topics in the field of immunothrombosis include platelets, inflammation, neutrophil extracellular traps, Von Willebrand factor, and the complement system. Research hotspots focus on the mechanisms and manifestations of immunothrombosis in COVID-19, as well as the discovery of novel treatment strategies targeting immunothrombosis in cardiovascular and cerebrovascular diseases. CONCLUSION Bibliometric analysis summarizes the main achievements and development trends in research on immunothrombosis, offering readers a comprehensive understanding of the field and guiding future research directions.
Collapse
Affiliation(s)
- Mengyu Hou
- Department of Research Ward, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Li S, Du Z, Ma H, Cai L, Liu X, He J. Mendelian randomization provides causal association between COVID-19 and thyroid cancer: insights from a multi-cancer analysis. Front Oncol 2024; 14:1419020. [PMID: 39319057 PMCID: PMC11419959 DOI: 10.3389/fonc.2024.1419020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/19/2024] [Indexed: 09/26/2024] Open
Abstract
Since the onset of the COVID-19 pandemic, the SARS-CoV-2 virus has caused over 600 million confirmed infections and more than 6.8 million deaths worldwide, with ongoing implications for human health. COVID-19 has been extensively documented to have extrapulmonary manifestations due to the widespread expression of necessary ACE2 receptors in the human body. Nevertheless, the association between COVID-19 and cancer risk remains inadequately explored. This study employs Mendelian randomization (MR) methods to examine the causal relationship between genetic variations associated with COVID-19 and the risk of developing cancer. The findings indicate that COVID-19 has negligible impact on most cancer risks. Interestingly, a higher COVID-19 impact is associated with a decreased risk of thyroid cancer. In summary, our findings demonstrate a genetic correlation between COVID-19 and thyroid cancer, contributing to our understanding of the interplay between COVID-19 and cancer risk.
Collapse
Affiliation(s)
- Shuhong Li
- Department of Oncology, Chengdu Second People’s Hospital, Chengdu, China
| | - Zedong Du
- Department of Oncology, Chengdu Second People’s Hospital, Chengdu, China
| | - Hui Ma
- Department of Oncology, Chengdu Second People’s Hospital, Chengdu, China
| | - Liang Cai
- Department of Oncology, Chengdu Second People’s Hospital, Chengdu, China
| | - Xiao Liu
- Department of Oncology, Chengdu Second People’s Hospital, Chengdu, China
| | - Jie He
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
39
|
Blasco A, Royuela A, García-Gómez S, Gómez-Lozano N, Sánchez-Arjona A, de la Fuente J, Anel J, Sánchez-Galarraga I, Pérez-Redondo M, González E, Silva L. Association of SARS-CoV-2 immunoserology and vaccination status with myocardial infarction severity and outcome. Vaccine 2024; 42:126305. [PMID: 39244425 DOI: 10.1016/j.vaccine.2024.126305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND The COVID-19 pandemic adversely affected the severity and prognosis of patients with acute myocardial infarction (MI) caused by atherothrombosis (type 1 MI). The effect, if any, of COVID-19 vaccination and natural SARS-CoV2 serologic immunity in these patients is unclear. Our aim was to analyze the association between the severity and outcome of patients with type 1 MI and their previous SARS-CoV2 vaccination and serostatus. METHODS A single-center retrospective cohort study conducted between March 1, 2020 and March 1, 2023. Clinical and follow-up information was collected from medical records and patients. Total antibodies (IgM, IgA, IgG) to nucleocapsid (N) antigens were measured by ECLIA (electrochemiluminescence-based immunoassay) to test the immune response to natural infection. If positive, IgM and IgG antibodies to spike (S) surface antigens were measured by CLIA to test the immune response to vaccine or natural infection. Multivariable logistic regression analysis was performed, adjusting for age, sex, hypertension, diabetes, and dyslipidemia. RESULTS Total sample of 949 patients, 656 with ST-segment elevation MI (STEMI) and 293 with non-ST-segment elevation MI (NSTEMI). Mean age was 64 (SD 13) years, 80 % men. Pre-admission vaccination status was: ≥ 1 dose, 53 % of patients; complete vaccination, 49 %; first booster dose, 25 %. The majority (84 %) of vaccines administered were mRNA-based. Six months after MI, 92 (9.7 %) patients had a major adverse cardiac event (MACE) and 50 died; 11 % of patients had severe heart failure or cardiogenic shock (Killip III-IV) after STEMI. Vaccinated patients with STEMI and positive serology (Pos/Vax group) had a higher risk of Killip III-IV on admission: OR 2.63 (1.27-5.44), p = 0.010. SARS-CoV-2 S-specific IgG titers were highest in this group (median > 2080 AU/mL, [IQR 1560- >2080] vs 91 [32-198] in the unvaccinated group). In the overall sample, a higher incidence of 6-month MACE was not demonstrated (OR 1.89 [0.98-3.61], p = 0.055). CONCLUSIONS The combination of vaccination and natural SARS-CoV2 infection was associated with the development of severe heart failure and cardiogenic shock in patients with STEMI, possibly related to an increased serological response.
Collapse
Affiliation(s)
- Ana Blasco
- Cardiology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain; Research Ethics Committee, Instituto de Investigación Puerta de Hierro-Segovia de Arana, Madrid, Spain.
| | - Ana Royuela
- Biostatistics Unit, Instituto de Investigación Puerta de Hierro-Segovia de Arana, Madrid, Spain; Center for Biomedical Research in Epidemiology and Public Health Network (CIBERESP), Madrid, Spain
| | - Sergio García-Gómez
- Cardiology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Natalia Gómez-Lozano
- Immunology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Alberto Sánchez-Arjona
- Cardiology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Jorge de la Fuente
- Cardiology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Jorge Anel
- Microbiology Department, Serology Section, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | | | - Marina Pérez-Redondo
- Intensive Care Unit, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Elisa González
- Cardiology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Lorenzo Silva
- Cardiology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| |
Collapse
|
40
|
di Filippo L, Franzese V, Santoro S, Doga M, Giustina A. Long COVID and pituitary dysfunctions: a bidirectional relationship? Pituitary 2024:10.1007/s11102-024-01442-8. [PMID: 39240511 DOI: 10.1007/s11102-024-01442-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/02/2024] [Indexed: 09/07/2024]
Abstract
Long COVID is a novel emerging syndrome known to affect multiple health areas in patients previously infected by SARS-CoV-2 markedly impairing their quality of life. The pathophysiology of Long COVID is still largely poorly understood and multiple mechanisms were proposed to underlie its occurrence, including alterations in the hormonal hypothalamic-pituitary axes. Aim of this review is to present and discuss the potential negative implications of these hormonal dysfunctions in promoting and influencing the Long COVID syndrome. To date, the hypothalamic-pituitary-adrenal axis is the mostly investigated and several studies have reported a prolonged impairment leading to mild and subclinical forms of central adrenal insufficiency. Few data are also available regarding central hypogonadism, central hypothyroidism and growth hormone (GH) deficiency. A high prevalence of central hypogonadism in COVID-19 survivors several months after recovery was consistently reported in different cohorts. Conversely, very few data are available on the hypothalamic-pituitary-thyroid axis function that was mainly shown to be preserved in COVID-19 survivors. Finally, a potential impairment of the hypothalamic-GH axis in Long COVID has also been reported. These data altogether may suggest a novel possible pituitary-centred pathophysiological view of Long COVID syndrome which if confirmed by large clinical studies may have relevant implication for the diagnostic and therapeutic approach at least in a subset of patients with the syndrome.
Collapse
Affiliation(s)
- Luigi di Filippo
- Institute of Endocrine and Metabolic Sciences, San Raffaele Vita-Salute University and IRCCS San Raffaele Hospital, Via Olgettina 60, Milan, 20132, Italy.
| | - Vincenzo Franzese
- Institute of Endocrine and Metabolic Sciences, San Raffaele Vita-Salute University and IRCCS San Raffaele Hospital, Via Olgettina 60, Milan, 20132, Italy
| | - Simona Santoro
- Institute of Endocrine and Metabolic Sciences, San Raffaele Vita-Salute University and IRCCS San Raffaele Hospital, Via Olgettina 60, Milan, 20132, Italy
| | - Mauro Doga
- Institute of Endocrine and Metabolic Sciences, San Raffaele Vita-Salute University and IRCCS San Raffaele Hospital, Via Olgettina 60, Milan, 20132, Italy
| | - Andrea Giustina
- Institute of Endocrine and Metabolic Sciences, San Raffaele Vita-Salute University and IRCCS San Raffaele Hospital, Via Olgettina 60, Milan, 20132, Italy
| |
Collapse
|
41
|
Feng SN, Kelly TL, Fraser JF, Li Bassi G, Suen J, Zaaqoq A, Griffee MJ, Arora RC, White N, Whitman G, Robba C, Battaglini D, Cho SM. Impact of Hemoglobin Levels on Composite Cardiac Arrest or Stroke Outcome in Patients With Respiratory Failure Due to COVID-19. Crit Care Explor 2024; 6:e1143. [PMID: 39172625 PMCID: PMC11343536 DOI: 10.1097/cce.0000000000001143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024] Open
Abstract
OBJECTIVES Anemia has been associated with an increased risk of both cardiac arrest and stroke, frequent complications of COVID-19. The effect of hemoglobin level at ICU admission on a composite outcome of cardiac arrest or stroke in an international cohort of COVID-19 patients was investigated. DESIGN Retrospective analysis of prospectively collected database. SETTING A registry of COVID-19 patients admitted to ICUs at over 370 international sites was reviewed for patients diagnosed with cardiac arrest or stroke up to 30 days after ICU admission. Anemia was defined as: normal (hemoglobin ≥ 12.0 g/dL for women, ≥ 13.5 g/dL for men), mild (hemoglobin 10.0-11.9 g/dL for women, 10.0-13.4 g/dL for men), moderate (hemoglobin ≥ 8.0 and < 10.0 g/dL for women and men), and severe (hemoglobin < 8.0 g/dL for women and men). PATIENTS Patients older than 18 years with acute COVID-19 infection in the ICU. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Of 6926 patients (median age = 59 yr, male = 65%), 760 patients (11.0%) experienced stroke (2.0%) and/or cardiac arrest (9.4%). Cardiac arrest or stroke was more common in patients with low hemoglobin, occurring in 12.8% of patients with normal hemoglobin, 13.3% of patients with mild anemia, and 16.7% of patients with moderate/severe anemia. Time to stroke or cardiac arrest by anemia status was analyzed using Cox proportional hazards regression with death as a competing risk. Covariates selected through clinical knowledge were age, sex, comorbidities (diabetes, hypertension, obesity, and cardiac or neurologic conditions), pandemic era, country income, mechanical ventilation, and extracorporeal membrane oxygenation. Moderate/severe anemia was associated with a higher risk of cardiac arrest or stroke (hazard ratio, 1.32; 95% CI, 1.05-1.67). CONCLUSIONS In an international registry of ICU patients with COVID-19, moderate/severe anemia was associated with increased hazard of cardiac arrest or stroke.
Collapse
Affiliation(s)
- Shi Nan Feng
- Division of Neuroscience Critical Care, Departments of Neurology, Neurosurgery, and Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Thu-Lan Kelly
- School of Public Health and Social Work, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - John F. Fraser
- Critical Care Research Group, Faculty of Medicine, Queensland University of Technology, Brisbane, QLD, Australia
- Adult Intensive Care Services, The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Gianluigi Li Bassi
- Critical Care Research Group, Faculty of Medicine, Queensland University of Technology, Brisbane, QLD, Australia
- Institut d’Investigacions Biomediques August Pi i Sunyer, Barcelona, Spain
| | - Jacky Suen
- Critical Care Research Group, Faculty of Medicine, Queensland University of Technology, Brisbane, QLD, Australia
| | - Akram Zaaqoq
- Department of Critical Care Medicine, MedStar Washington Hospital Center, Georgetown University, Washington, DC
| | - Matthew J. Griffee
- Department of Anesthesiology, University of Utah School of Medicine, Salt Lake City, UT
| | - Rakesh C. Arora
- Harrington Heart and Vascular Institute, University Hospitals, Cleveland, OH
- Department of Surgery, Case Western Reserve University, Cleveland, OH
| | - Nicole White
- School of Public Health and Social Work, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
- Critical Care Research Group, Faculty of Medicine, Queensland University of Technology, Brisbane, QLD, Australia
| | - Glenn Whitman
- Division of Neuroscience Critical Care, Departments of Neurology, Neurosurgery, and Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Chiara Robba
- School of Public Health and Social Work, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Denise Battaglini
- School of Public Health and Social Work, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Sung-Min Cho
- Division of Neuroscience Critical Care, Departments of Neurology, Neurosurgery, and Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
42
|
Bayram N, Ozsaygılı C, Gundogan M, Unal S, Bagci F, Pangal E, Yuvacı IS. The Impact of Severe COVID-19 on Corneal Endothelial Cells-Analysis of the In Vivo Noncontact Specular Microscopy. Ocul Immunol Inflamm 2024; 32:1228-1236. [PMID: 37348068 DOI: 10.1080/09273948.2023.2219312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 03/06/2023] [Accepted: 05/24/2023] [Indexed: 06/24/2023]
Abstract
PURPOSE This study aimed to investigate the effects of severe COVID-19 infection on the corneal endothelium via in vivo specular microscopy. METHODS This was an observational, prospective, and controlled study including 56 eyes of 56 severe COVID-19 patients, compared to after-recovery and 56 eyes of 56 age- and gender-matched healthy controls. RESULTS Endothelial cell density was lower in the active disease period compared to healthy controls (p = .001) and decreased even more after recovery (p < .0001). After recovery, the average cell area and coefficient of variation were higher compared to the active disease period (p < .0001 and p = .008, respectively) and the healthy controls (for both, p < .0001), whereas hexagonality was lower (p < .0001). Central corneal thickness increased in the active disease period compared to after recovery (p < .0001) and healthy controls (p = .002). CONCLUSIONS These results may be due to direct host-virus interaction or linked to immune dysregulation, subclinical corneal endotheliitis, or still yet a viral-mediated inflammation.
Collapse
Affiliation(s)
- Nurettin Bayram
- Kayseri City Training and Research Hospital, Department of Ophthalmology, University of Health Science, Kayseri, Turkey
- Etlik City Training and Research Hospital, Department of Ophthalmology, University of Health Sciences, Ankara, Turkey
| | - Cemal Ozsaygılı
- Kayseri City Training and Research Hospital, Department of Ophthalmology, University of Health Science, Kayseri, Turkey
| | - Medine Gundogan
- Kayseri City Training and Research Hospital, Department of Ophthalmology, University of Health Science, Kayseri, Turkey
| | - Sefa Unal
- Kayseri City Training and Research Hospital, Department of Ophthalmology, University of Health Science, Kayseri, Turkey
| | - Fatma Bagci
- Etlik City Training and Research Hospital, Department of Ophthalmology, University of Health Sciences, Ankara, Turkey
| | - Emine Pangal
- Kayseri City Training and Research Hospital, Department of Ophthalmology, University of Health Science, Kayseri, Turkey
| | - I Sa Yuvacı
- Department of Ophthalmology, Sakarya University Medical Education and Research Hospital, Sakarya, Turkey
| |
Collapse
|
43
|
Zhang L, Qin J, Li P. Bioinformatics analysis of potential common pathogenic mechanisms for COVID-19 and venous thromboembolism. Cytokine 2024; 181:156682. [PMID: 38909539 DOI: 10.1016/j.cyto.2024.156682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/20/2024] [Accepted: 06/18/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND A growing body of research has shown that patients with coronavirus disease 2019 (COVID-19) have significantly higher rates of venous thromboembolism (VTE) than healthy. However, the mechanism remains incompletely elucidated. This study aimed to further investigate the molecular mechanisms underlying the development of this complication. METHODS The gene expression profiles of COVID-19 and VTE were downloaded from the Gene Expression Omnibus (GEO) database. After identifying the common differentially expressed genes (DEGs) for COVID-19 and VTE, functional annotation, a protein-protein interactions (PPI) network, module construction, and hub gene identification were performed. Finally, we constructed a transcription factor (TF)-gene regulatory network and a TF-miRNA regulatory network for hub genes. RESULTS A total of 42 common DEGs were selected for subsequent analyses. Functional analyses showed that biological function and signaling pathways collectively participated in the development and progression of VTE and COVID-19. Finally, 8 significant hub genes were identified using the cytoHubba plugin, including RSL24D1, RPS17, RPS27, HINT1, COX7C, RPL35, RPL34, and NDUFA4, which had preferable values as diagnostic markers for COVID-19 and VTE. CONCLUSIONS Our study revealed the common pathogenesis of COVID-19 and VTE. These common pathways and pivotal genes may provide new ideas for further mechanistic studies.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Jing Qin
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Peiwu Li
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, Gansu, China.
| |
Collapse
|
44
|
Steinbeis F, Kedor C, Meyer HJ, Thibeault C, Mittermaier M, Knape P, Ahrens K, Rotter G, Temmesfeld-Wollbrück B, Sander LE, Kurth F, Witzenrath M, Scheibenbogen C, Zoller T. A new phenotype of patients with post-COVID-19 condition is characterised by a pattern of complex ventilatory dysfunction, neuromuscular disturbance and fatigue symptoms. ERJ Open Res 2024; 10:01027-2023. [PMID: 39377086 PMCID: PMC11456967 DOI: 10.1183/23120541.01027-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 05/10/2024] [Indexed: 10/09/2024] Open
Abstract
Background Patients with post-COVID-19 condition frequently suffer from chronic dyspnoea. The causes and mechanism for dyspnoea in these patients without evidence of structural lung disease are unclear. Methods Patients treated for COVID-19 at Charité University Hospital in Berlin received pulmonary function testing including respiratory muscle strength tests and completed health-related quality-of-life questionnaires during follow-up. Patients with post-COVID-19 condition during outpatient follow-up with fatigue and exertional intolerance (PCF) were compared to patients with post-COVID-19 condition with evidence of chronic pulmonary sequelae (post-COVID-19 restriction (PCR)) as well as to patients without post-COVID-19 condition (NCF). Results A total of 170 patients presented for follow-up. 36 participants met criteria for PCF, 28 for PCR and 24 for NCF. PCF patients reported dyspnoea in 63.8%. % predicted value of respiratory muscle strength (median (IQR)) was reduced in PCF (55.8 (41.5-75.9)) compared to NCF and PCR (70.6 (66.3-88.9) and 76.8 (63.6-102.2), respectively; p=0.011). A pattern of reduced forced vital capacity (FVC), but normal total lung capacity (TLC), termed complex ventilatory dysfunction defined as TLC - FVC >10% predicted was observed and occurred more frequently in PCF (88.9%) compared to NCF and PCR (29.1% and 25.0%, respectively; p<0.001). Conclusion Dyspnoea in PCF is characterised by reduced respiratory muscle strength and complex ventilatory dysfunction indicating neuromuscular disturbance as a distinct phenotype among patients with post-COVID-19 condition. These observations could be a starting point for developing personalised rehabilitation concepts.
Collapse
Affiliation(s)
- Fridolin Steinbeis
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
| | - Claudia Kedor
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Medical Immunology, Berlin, Germany
| | - Hans-Jakob Meyer
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
| | - Charlotte Thibeault
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Mirja Mittermaier
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Philipp Knape
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
| | - Katharina Ahrens
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
| | - Gabriele Rotter
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Social Medicine, Epidemiology and Health Economics, Berlin, Germany
| | - Bettina Temmesfeld-Wollbrück
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
| | - Leif Erik Sander
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
- German Center for Lung Research (DZL), Berlin, Germany
| | - Florian Kurth
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
- Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Department of Medicine I, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Witzenrath
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
- German Center for Lung Research (DZL), Berlin, Germany
| | - Carmen Scheibenbogen
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Medical Immunology, Berlin, Germany
| | - Thomas Zoller
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
| |
Collapse
|
45
|
Iness AN, Shah KM, Kukreja RC. Physiological effects of ivabradine in heart failure and beyond. Mol Cell Biochem 2024; 479:2405-2414. [PMID: 37768496 DOI: 10.1007/s11010-023-04862-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023]
Abstract
Ivabradine is a pharmacologic agent that inhibits the funny current responsible for determining heart rate in the sinoatrial node. Ivabradine's clinical potential has been investigated in the context of heart failure since it is associated with reduced myocardial oxygen demand, enhanced diastolic filling, stroke volume, and coronary perfusion time; however, it is yet to demonstrate definitive mortality benefit. Alternative effects of ivabradine include modulation of the renin-angiotensin-aldosterone system, sympathetic activation, and endothelial function. Here, we review key clinical trials informing the clinical use of ivabradine and explore opportunities for leveraging its potential pleiotropic effects in other diseases, including treatment of hyperadrenergic states and mitigating complications of COVID-19 infection.
Collapse
Affiliation(s)
- Audra N Iness
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Keyur M Shah
- Division of Cardiology, Pauley Heart Center, Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Rakesh C Kukreja
- Division of Cardiology, Pauley Heart Center, Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
46
|
Class J, Simons LM, Lorenzo-Redondo R, Achi JG, Cooper L, Dangi T, Penaloza-MacMaster P, Ozer EA, Lutz SE, Rong L, Hultquist JF, Richner JM. Evolution of SARS-CoV-2 in the murine central nervous system drives viral diversification. Nat Microbiol 2024; 9:2383-2394. [PMID: 39179693 DOI: 10.1038/s41564-024-01786-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 07/18/2024] [Indexed: 08/26/2024]
Abstract
Severe coronavirus disease 2019 and post-acute sequelae of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are associated with neurological complications that may be linked to direct infection of the central nervous system (CNS), but the selective pressures ruling neuroinvasion are poorly defined. Here we assessed SARS-CoV-2 evolution in the lung versus CNS of infected mice. Higher levels of viral divergence were observed in the CNS than the lung after intranasal challenge with a high frequency of mutations in the spike furin cleavage site (FCS). Deletion of the FCS significantly attenuated virulence after intranasal challenge, with lower viral titres and decreased morbidity compared with the wild-type virus. Intracranial inoculation of the FCS-deleted virus, however, was sufficient to restore virulence. After intracranial inoculation, both viruses established infection in the lung, but dissemination from the CNS to the lung required the intact FCS. Cumulatively, these data suggest a critical role for the FCS in determining SARS-CoV-2 tropism and compartmentalization.
Collapse
Affiliation(s)
- Jacob Class
- Department of Microbiology and Immunology, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Lacy M Simons
- Department of Medicine, Division of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Havey Institute for Global Health, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ramon Lorenzo-Redondo
- Department of Medicine, Division of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Havey Institute for Global Health, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jazmin Galván Achi
- Department of Microbiology and Immunology, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Laura Cooper
- Department of Microbiology and Immunology, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Tanushree Dangi
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Pablo Penaloza-MacMaster
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Egon A Ozer
- Department of Medicine, Division of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Havey Institute for Global Health, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Sarah E Lutz
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Lijun Rong
- Department of Microbiology and Immunology, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Judd F Hultquist
- Department of Medicine, Division of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Center for Pathogen Genomics and Microbial Evolution, Havey Institute for Global Health, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | - Justin M Richner
- Department of Microbiology and Immunology, College of Medicine, University of Illinois Chicago, Chicago, IL, USA.
| |
Collapse
|
47
|
Wu K, Van Name J, Xi L. Cardiovascular abnormalities of long-COVID syndrome: Pathogenic basis and potential strategy for treatment and rehabilitation. SPORTS MEDICINE AND HEALTH SCIENCE 2024; 6:221-231. [PMID: 39234483 PMCID: PMC11369840 DOI: 10.1016/j.smhs.2024.03.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 03/13/2024] [Accepted: 03/19/2024] [Indexed: 09/06/2024] Open
Abstract
Cardiac injury and sustained cardiovascular abnormalities in long-COVID syndrome, i.e. post-acute sequelae of coronavirus disease 2019 (COVID-19) have emerged as a debilitating health burden that has posed challenges for management of pre-existing cardiovascular conditions and other associated chronic comorbidities in the most vulnerable group of patients recovered from acute COVID-19. A clear and evidence-based guideline for treating cardiac issues of long-COVID syndrome is still lacking. In this review, we have summarized the common cardiac symptoms reported in the months after acute COVID-19 illness and further evaluated the possible pathogenic factors underlying the pathophysiology process of long-COVID. The mechanistic understanding of how Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) damages the heart and vasculatures is critical in developing targeted therapy and preventive measures for limiting the viral attacks. Despite the currently available therapeutic interventions, a considerable portion of patients recovered from severe COVID-19 have reported a reduced functional reserve due to deconditioning. Therefore, a rigorous and comprehensive cardiac rehabilitation program with individualized exercise protocols would be instrumental for the patients with long-COVID to regain the physical fitness levels comparable to their pre-illness baseline.
Collapse
Affiliation(s)
- Kainuo Wu
- Virginia Commonwealth University School of Medicine (M.D. Class 2024), Richmond, VA, 23298, USA
| | - Jonathan Van Name
- Virginia Commonwealth University School of Medicine (M.D. Class 2024), Richmond, VA, 23298, USA
| | - Lei Xi
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, 23298-0204, USA
| |
Collapse
|
48
|
Singh H, Singh S, Sadhar B, Winegarden B, Morrissey S. Exploring COVID-19-Associated Ileus: A Compelling Case Study. Cureus 2024; 16:e69647. [PMID: 39429337 PMCID: PMC11488154 DOI: 10.7759/cureus.69647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2024] [Indexed: 10/22/2024] Open
Abstract
Since the start of 2020, COVID-19 has profoundly impacted global health. Classically, the disease manifests with pulmonary symptoms such as cough, dyspnea, pneumonia, and respiratory distress. However, it is important to note that in addition to these pulmonary symptoms, COVID-19 can present with extrapulmonary symptoms. This case report involves an 82-year-old male who tested positive for the causative agent of COVID-19 - severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), whose presentation manifested with gastrointestinal symptoms in the form of colonic ileus. This report aims to highlight the pathophysiological mechanisms of COVID-19-associated ileus while also delving into the importance of timely intervention to prevent complications like intestinal perforation.
Collapse
Affiliation(s)
- Harpreet Singh
- Medicine, Lake Erie College of Osteopathic Medicine, Erie, USA
- Surgery, Conemaugh Memorial Medical Center, Johnstown, USA
| | - Satnam Singh
- Medicine, Lake Erie College of Osteopathic Medicine, Erie, USA
- Surgery, Conemaugh Memorial Medical Center, Johnstown, USA
| | - Birkaran Sadhar
- Medicine, Lake Erie College of Osteopathic Medicine, Erie, USA
- Surgery, Conemaugh Memorial Medical Center, Johnstown, USA
| | | | - Shawna Morrissey
- Trauma and Acute Care Surgery, Conemaugh Memorial Medical Center, Johnstown, USA
| |
Collapse
|
49
|
Liu AQ, Choy EKH, Chiu PKF, Yee CH, Ng CF, Teoh JYC. High Levels of Incidental COVID-19 Infection in Emergency Urology Admissions: A Propensity Score-Matched Real World Data Analysis across Surgical Specialties. Viruses 2024; 16:1402. [PMID: 39339878 PMCID: PMC11437490 DOI: 10.3390/v16091402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/22/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024] Open
Abstract
Background: An incidental COVID-19 infection is often found in patients admitted for non-COVID-19-related conditions. This study aims to investigate the incidence of COVID-19 infections across surgical specialties including urology, general surgery, and orthopaedic surgery. Methods: This is a retrospective cohort study based on a territory-wide electronic database in Hong Kong. All emergency in-hospital admissions under the urology, general surgery, and orthopaedic surgery divisions in the public healthcare system in Hong Kong from January to September 2022 were included. All patients were routinely screened for SARS-CoV-2, based on admission protocols during the investigation period. Baseline characteristics were retrieved, with 1:1:1 propensity score matching being performed. Incidental COVID-19 rates were then compared across specialties. Results: A total of 126,034 patients were included. After propensity score matching, the baseline characteristics were well balanced, and 8535 patients in each group were analysed. Urology admission was noted to have a statistically significant higher incidence of incidental COVID-19 at 9.3%, compared to general surgery (5.4%) or orthopaedic surgery (5.6%). Amongst urology patients with incidental COVID-19 infection, 35.8% were admitted for retention of urine, 27.9% for haematuria, and 8.6% for a urinary tract infection. Conclusions: This large-scale cohort study demonstrated that incidental COVID-19 rates differ between surgical specialties, with urology having the highest proportion of incidental COVID-19 infection.
Collapse
Affiliation(s)
- Alex Qinyang Liu
- S.H. Ho Urology Centre, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China; (E.K.-H.C.); (P.K.-F.C.); (C.-H.Y.); (C.-F.N.)
| | - Eric Ka-Ho Choy
- S.H. Ho Urology Centre, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China; (E.K.-H.C.); (P.K.-F.C.); (C.-H.Y.); (C.-F.N.)
| | - Peter Ka-Fung Chiu
- S.H. Ho Urology Centre, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China; (E.K.-H.C.); (P.K.-F.C.); (C.-H.Y.); (C.-F.N.)
| | - Chi-Hang Yee
- S.H. Ho Urology Centre, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China; (E.K.-H.C.); (P.K.-F.C.); (C.-H.Y.); (C.-F.N.)
| | - Chi-Fai Ng
- S.H. Ho Urology Centre, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China; (E.K.-H.C.); (P.K.-F.C.); (C.-H.Y.); (C.-F.N.)
| | - Jeremy Yuen-Chun Teoh
- S.H. Ho Urology Centre, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China; (E.K.-H.C.); (P.K.-F.C.); (C.-H.Y.); (C.-F.N.)
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Department of Urology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
50
|
Ceja-Gálvez HR, Hernández-Ramírez CO, Vega-Magaña AN, Hernández-Bello J, Arellano-Arteaga KJ, Turrubiates-Hernández FJ, Padilla-Borquez DL, Muñoz-Valle JF. Clinical characteristics and severity markers in hospitalized COVID-19 patients from western Mexico: a comparative analysis of Delta and Omicron variants. Front Public Health 2024; 12:1425372. [PMID: 39281077 PMCID: PMC11392767 DOI: 10.3389/fpubh.2024.1425372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/12/2024] [Indexed: 09/18/2024] Open
Abstract
Introduction COVID-19 is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a virus notable for its rapid mutation rate, which has led to the emergence of various variants such as Delta and Omicron, each with potentially different levels of transmissibility and virulence. Therefore, this study aims to compare clinical charactheristics and markers associated with the severity of COVID-19 in hospitalized patients from western Mexico who were infected with the Delta and Omicron variants of SARS-CoV-2. Methods This cross-sectional study involved 66 patients hospitalized for COVID-19, diagnosed by RT-qPCR. SARS-CoV-2 variants were identified through whole genome sequencing using the COVIDseq platform from Illumina. Upon admission, patients underwent a clinical history assessment, blood gas analysis, and blood biometry. Additionally, several tests and markers were measured, including the percentage of neutralizing antibodies, erythrocyte sedimentation rate (ESR), interleukin-6 (IL-6), tumor necrosis factor-alpha (TNFα), D-dimer, lactate dehydrogenase (LDH), C-reactive protein (CRP), and ferritin. Results and discussion Patients hospitalized with the Omicron were found to be older, compared to those infected with the Delta (64 vs. 54 years, p = 0.006). Additionally, a higher proportion of male patients were observed in the Omicron compared to the Delta (p = 0.029). Both Omicron and Delta variants were associated with lymphopenia, although the lymphocyte count was lower in Omicron (0.9 vs. 0.56 10x3/L; p = 0.007). The COVID-GRAM scale indicated a high risk for severe disease in both groups, but the score was higher in Omicron compared to Delta (157 vs. 128 points; p = 0.0004). Patients infected with Omicron exhibited a lower percentage of neutralizing antibodies than those with Delta (35.99 vs. 81%; p < 0.05), regardless of their vaccination status. Among the markers assessed, globular ESR was found to be lower in Omicron compared to Delta (30.5 vs. 41.5 mm/h; p = 0.001), while ferritin levels were higher in patients infected with the Omicron (1,359 vs. 960.6 μg/L; p = 0.007). In patients with severe COVID-19, markers such as lymphopenia, neutralizing antibody levels, ferritin, and COVID-GRAM scores are elevated in the Omicron variant, while only the leukocyte count and ESR for the Delta variant.
Collapse
Affiliation(s)
- Hazael Ramiro Ceja-Gálvez
- Institute of Research in Biomedical Sciences, Centro Universitario de Ciencias de la Salud (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Cristian Oswaldo Hernández-Ramírez
- Institute of Research in Biomedical Sciences, Centro Universitario de Ciencias de la Salud (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Alejandra Natali Vega-Magaña
- Institute of Research in Biomedical Sciences, Centro Universitario de Ciencias de la Salud (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Jorge Hernández-Bello
- Institute of Research in Biomedical Sciences, Centro Universitario de Ciencias de la Salud (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
| | | | - Francisco Javier Turrubiates-Hernández
- Institute of Research in Biomedical Sciences, Centro Universitario de Ciencias de la Salud (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Diana Lourdes Padilla-Borquez
- Institute of Research in Biomedical Sciences, Centro Universitario de Ciencias de la Salud (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - José Francisco Muñoz-Valle
- Institute of Research in Biomedical Sciences, Centro Universitario de Ciencias de la Salud (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
| |
Collapse
|